WO2014098877A1 - Substituted noribogaine - Google Patents

Substituted noribogaine Download PDF

Info

Publication number
WO2014098877A1
WO2014098877A1 PCT/US2012/071052 US2012071052W WO2014098877A1 WO 2014098877 A1 WO2014098877 A1 WO 2014098877A1 US 2012071052 W US2012071052 W US 2012071052W WO 2014098877 A1 WO2014098877 A1 WO 2014098877A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
optionally substituted
hydrogen
group
alkyl
Prior art date
Application number
PCT/US2012/071052
Other languages
French (fr)
Inventor
Deborah Mash
Richard D. Gless
Robert M. Moriarty
Original Assignee
Demerx, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Demerx, Inc. filed Critical Demerx, Inc.
Priority to CA2896133A priority Critical patent/CA2896133A1/en
Priority to JP2015549336A priority patent/JP2016508979A/en
Priority to US14/654,503 priority patent/US9783535B2/en
Priority to PCT/US2012/071052 priority patent/WO2014098877A1/en
Priority to EP12890228.5A priority patent/EP2934541A4/en
Priority to US13/732,751 priority patent/US8940728B2/en
Priority to US13/753,364 priority patent/US9045481B2/en
Publication of WO2014098877A1 publication Critical patent/WO2014098877A1/en
Priority to US14/719,041 priority patent/US20150361080A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D453/00Heterocyclic compounds containing quinuclidine or iso-quinuclidine ring systems, e.g. quinine alkaloids
    • C07D453/06Heterocyclic compounds containing quinuclidine or iso-quinuclidine ring systems, e.g. quinine alkaloids containing isoquinuclidine ring systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/47Quinolines; Isoquinolines
    • A61K31/485Morphinan derivatives, e.g. morphine, codeine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/55Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/04Centrally acting analgesics, e.g. opioids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/30Drugs for disorders of the nervous system for treating abuse or dependence
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/30Drugs for disorders of the nervous system for treating abuse or dependence
    • A61P25/32Alcohol-abuse
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/30Drugs for disorders of the nervous system for treating abuse or dependence
    • A61P25/34Tobacco-abuse
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P39/00General protective or antinoxious agents
    • A61P39/02Antidotes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/22Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed systems contains four or more hetero rings

Definitions

  • This invention relates noribogaine derivatives, compositions and methods of use thereof.
  • Noribogaine is a well known derivative of ibogaine and is sometimes referred to as 12-hydiOxyibogaine. It is a metabolite of ibogaine.
  • the structure of noribogaine has now been thoroughly evaluated and is found to combine the features of tryptamine, tetrahydrohavaine and indolazepines.
  • Noribogaine can be depicted by the following formula:
  • Noribogaine and its pharmaceutically acceptable salts have recently received significant attention as a non-addictive alkaloid useful in treating addiction (U.S. Patent No. 6.348,456) and as a potent analgesic (U.S. Patent No. 7,220,737).
  • Noribogaine has been found to have properties suitable for the treatment of pain and to the withdrawal symptoms associated with drug dependency or abuse.
  • noribogaine binds to two classes of opioid receptors that have been associated with pain relief, the ⁇ and ⁇ receptors.
  • noribogaine acts as an opiate agonist.
  • noribogaine elevates brain serotonin levels by blocking synaptic reuptake. It is believed that such levels (as well as ligand interactions at the ⁇ and ⁇ opiate receptors) play a role in the anxiety and drug cravings experienced by addicts during withdrawal,
  • Noribogaine analogs are also found to inhibit a3b4 nicotinic acetylcholine receptors (nAChRs).
  • Noribogaine analogs are also found effective for treatment of nicotine addiction and for treatment of other substance abuse related disorders (SRDs). Summary of the Invention
  • This invention relates to noribogaine derivative compounds. Such compounds are contemplated as being useful in treating drug addiction and/or pain. Accordingly, in one of its compound aspects, this invention is directed to a compound or a pharmaceutically acceptable salt thereof wherein said compound is represented by Formula IA or Formula IB, or a pharmaceutically acceptable salt of each thereof:
  • R is OR or Ci-C] 2 alkyl optionally substituted with 1 to 5 R ;
  • R 1 is selected from the group consisting of hydrogen, -C(0)OX, -S0 2 OR , 1'0. a
  • X is Ci -C 6 alkyl optionally substituted with 1 to 5 R 9
  • each Y is independently selected from the group consisting of hydrogen, C)-C6 alkyl optionally substituted with 1 to 5 R 9 , C 6 -C] 4 aryl optionally substituted with 1 to 5 R 9 , C3-C 10 cycloalkyl optionally substituted with 1 to 5 R 9 , Cj-Ci o heteroaryl having 1 to 4 heteroatoms and which is optionally substituted with 1 to 5 R 9 , d-C 10 heterocyclic having 1 to 4 heteroatoms and which is optionally substituted with 1 to 5 R 9 .
  • each Y. together with the nitrogen atom bound thereto form either a C 1 -C 6 heterocyclic having 1 to 4 heteroatoms and which is optional ly substituted with 1 to 5 R 9 , or a Ci-Cg heteroaryl having 1 to 4 heteroatoms and which is optionally substituted with 1 to 5 R 9 ;
  • R' is halo. OR 20 , or Ci-C 12 alkyl optionally substituted with 1 to 5 R 9 ;
  • R 2 is selected from the group consisting of hydrogen . -C(0)X, -C(0)OX and -C(0)N Y ) 2 where X and Y are defined as above;
  • R 2 is hydrogen, -SO 2 OR i . a monophosphate, a diphosphate, or a triphosphate;
  • R J is selected from the group consisting of hydrogen, -(CFl 2 ) m OR ', -CR 6 (()H)R 7 , -(CH 2 ) m CN, -(CH 2 )mCOR 7 , -(CH 2 ) m C0 2 R 7 , -(CH 2 ) m C(0)NR 6 R 7 , ⁇ (CFI 2 ) m C(0)NR 6 NR 7 R 7 , m is 0, 1 , or 2;
  • L is a bond or C 1 -C 12 alkylene
  • R 4 is selected from the group consisting of C 1 -C 12 alkyl substituted with 1 to 5 R 9 , C2-C12 alkenyl substituted with 1 to 5 R 9 , -X 2 -R 6 , -(X 2 -Y 2 ) n -X 2 -R 6 , -S0 2 NR 6 R 7 , -0-C(0)R 8 , -C(0)OR 7 , -C(0)NR 6 R 7 , -NR 6 R ⁇ -NHC(0)R 8 , and -NR 6 C(0)R 8 ;
  • X 2 is selected from the group consisting of O and S;
  • Y 2 is C 1-C4 alkylene or QJ-C IQ arylene, or a combination thereof;
  • n 1, 2, or 3;
  • R 6 and R' are each independently selected from the group consisting of hydrogen, C 1 -C 12 alkyl optionally substituted with 1 to 5 R 9 , C1 -C6 heterocycle having 1 to 4 heteroatoms which is optionally substituted with 1 to 5 R 9 , C3-Cio cycioalkyl optionally substituted with 1 to 5 R 9 , Ce-Cio aryl optionally substituted with 1 to 5 R 9 and Ci- heteroaryl having 1 to 4 heteroatoms optionally substituted with 1 to 5 R 9 ; or
  • R is selected from the group consisting of C
  • R 9 is selected from the group consisting of C1 -C4 alkyl, phenyl, halo, -OH, -OR 10 , -CN,
  • R 10 is C1-C12 alkyl
  • R 3 is hydrogen
  • This invention is also directed to pharmaceutical compositions comprising a pharmaceutically acceptable excipient and a therapeutically effective amount of a compound of this invention or a mixture of one or more of such compounds.
  • This invention is still further directed to methods for treating pain and/or addiction in a patient in need thereof, which methods comprise administering to the patient a one or more of compounds or a pharmaceutical composition of this invention.
  • This invention is also directed to methods of inhibiting a3b4 nicotinic acetylcholine receptors.
  • This invention is also directed to methods for treating of nicotine addiction and methods for treating of other substance abuse related disorders (SRDs ).
  • SRDs substance abuse related disorders
  • This invention is directed to ribose substituted noribogaine for use in treating pain and/or addition.
  • ribose substituted noribogaine for use in treating pain and/or addition.
  • compositions and methods are intended to mean that the compositions and methods include the recited elements, but not excluding others.
  • compositions and methods shall mean excluding other elements of any essential significance to the combination for the stated purpose. Thus, a composition consisting essentially of the elements as defined herein would not exclude other materials or steps that do not materially affect the basic and novel characteristic(s) of the claimed invention. "Consisting of shall mean excluding more than trace elements of other ingredients and substantial method steps. Embodiments defined by each of these transition terms are within the scope of this invention.
  • Noribogaine can be depicted by the following Formula:
  • Ibogaine may be obtained from natural or commercial sources, or can be synthesized by methods known in the art (see Huffman, et al, J. Org. Chem. 50: 1460 (1 85)).
  • noribogaine may also be obtained from the National Institute on Drug Abuse (Rockville, d.j.
  • hydrolyzable group 1 ' refers to a group that can be hydrolyzed to release the free hydroxy group under hydrolysis conditions.
  • hydrolvsable group include, but are not limited to those defined for R 1 above.
  • Preferred hydrolvsable groups include carboxyl esters, phosphates and phosphate esters.
  • the hydrolysis may be done by chemical reactions conditions such as base hydrolysis or acid hydrolysis or may be done in vivo by biological processes, such as those catalyzed by a phosphate hydrolysis enzyme
  • hydrolysable group include groups linked with an cstcr-bascd linker (-C(O)O- or -OC(U)-), an amide-based linker (-C(O)NR 40 - or -NR 40 C(O)-), or a phosphate-linker (-P(O)(OR 40 )-O-, -O-P(S)(OR 40 )-O-.
  • Alkyl refers to alkyl groups having from 1 to 12 carbon atoms, preferably 1 to 6 carbon atoms and more preferably 1 to 3 carbon atoms.
  • the alkyl group may contain linear or branched carbon chains. This term is exemplified by groups such as methyl, ethyl, n- propyl, iso-propyl, n-butyl, L-butyl. n-pentyl and the like.
  • C x alkyl refers to an alkyl group having x carbon atoms, wherein x is an integer, for example.
  • C 3 refers to an alkyl group having 3 carbon atoms.
  • groups are exemplified, for example, by vinyl, allyl, and but-3-en- l -yl. Included within this term are the cis and trans isomers or mixtures of these isomers.
  • Alkynyl refers to straight or branched monovalent hydrocarbyl groups having from 2 to 12 carbon atoms, preferably 2 to 6 carbon atoms and more preferably 2 to 3 carbon atoms and having at least 1 and preferably from 1 to 2 sites of acetylenic (-C ⁇ C-) unsatiiration. Examples of such alkynyl groups include acetylenyl (-C ⁇ CH), and propargyl
  • Alkoxy refers to the group -O-alkyl wherein alkyl is as defined herein. Alkoxy includes, by way of example, methoxy, ethoxy, n-propoxy, iso-propoxy, n-butoxy, t-butoxy, sec-butoxy, n-pentoxy and the like.
  • AevT * refers to the groups R 4 l -C(0)-, wherein R 41 is selected from the group consisting of hydrogen, aikyl, cycloalkyl. aryl, heteroaryl, and heterocyclic, wherein alkyl. cycloalkyl, aryl, heteroaryl, and heterocyclic are as defined herein and are optionally substituted with 1 to 5 substituents independently selected from the group consisting of halo, Ci to C 3 alkyl, hydroxy!, and C( to C 3 alkoxy.
  • Aryl refers to a monovalent aromatic carbocyclic group of from 6 to 14 carbon atoms having a single ring (e.g., phenyl) or multiple condensed rings (e.g., naphthyl or anthryl) which condensed rings may or may not be aromatic (e.g., 2-benzoxazoIinone, 2H- l ,4-bcnzoxazin-3(4H)-one-7-yl, and the like) provided that the point of attachment is at an aromatic carbon atom.
  • Aryloxy refers to the group -O-aryl wherein aryl is as defined herein.
  • Aryloxy includes, by way of example, methoxy, ethoxy, n-propoxy. iso-propoxy, n-butoxy. t-butoxy, sec-butoxy, n-pentoxy and the like.
  • Carboxyl refers to -COOFI or salts thereof.
  • Carboxyl esters refers to the groups -C(0)0-R 42 , wherein R 42 is selected from the group consisting of hydrogen, alkyl, cycloalkyl, aryl, heteroaryl, and heterocyclic, wherein alkyl, cycloalkyl, aryl, heteroaryl, and heterocyclic are as defined herein and are optionally substituted w ith 1 to 5 substituents independently selected from the group consisting of halo, C ] to C 3 alkyl, hydroxy!, and C
  • Cycloalkyl refers to cyclic alkyl groups of from 3 to 10 carbon atoms having single or multiple cyclic rings including, by way of example, adamantyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclooctyl and the like.
  • Halo or "halogen” refers to fluoro, chloro, bromo and iodo and preferably is fluoro or chloro.
  • Heteroaryl refers to an aromatic group of from 1 to 10 carbon atoms and 1 to 4 heteroatoms selected from the group consisting of oxygen, nitrogen, sulfur within the ring, wherein the nitrogen and/or sulfur atom(s) of the heteroaryl are optionally oxidized (e.g., N- oxide, ⁇ S(0)- or -S( ( > >_- >.
  • Such heteroaryl groups can have a single ring (e.g., pyridyl or furyl) or multiple condensed rings (e.g..
  • indolizinyl or benzothienyl wherein the condensed rings may or may not be aromatic and/or contain a heteroatom provided that the point of attachment is through an atom of the aromatic heteroaryl group.
  • heteroaryls include pyridyl, pyrrolyl, indolyl, thiophenyl, and furyl,
  • Ielerocycle or “heterocyclic ' "' or “heterocycloalky” or “heterocyclyl” refers to a saturated or partially saturated, but not aromatic, group having from 1 to 10 ring carbon atoms and from 1 to 4 ring heteroatoms selected from the group consisting of nitrogen, sulfur, or oxygen. Heterocycle encompasses single ring or multiple condensed rings, including fused bridged and spiro ring systems. In fused ring systems, one or more the rings can be cycloalkyl, ar l, or heteroaryl provided that the point of attachment is through the
  • the nitrogen and/or sulfur atom(s) of the heterocyclic group arc optionally oxidized to provide for the N -oxide, sulfinyl, and/or sulfonyl moieties.
  • heteroeycles and heteroaryls include, but are not limited to, azetidine, pyrrole, imidazole, pyrazole, pyridine, pyrazine, pyrimidine, pyridazine, indolizinc, isoindole, indole, dihydroindole, indazole, purine, quinolizine, isoquinoline, quinoline, phthalazine, naphthylpyridine, quinoxaline, quinazoline, cinnoline. pteridine. carbazole, carboline, phenanthridine. acridine. phenanthroline.
  • morpholinyl, thiomorpholinyl also referred to as
  • thiamorpholinyl piperidinyl, pyrrolidine, tetrahydrofuranyl, and the like.
  • Alkylene refers to a divalent radical of an alkyl group.
  • ' Methylene refers to the group -CH 2 -.
  • Arylcnc refers to a divalent radical of an aryl group.
  • Phenylene refers to the divalent phenyl group - ( ' ⁇ ,! I >,-.
  • phosphate ester ' refers to any one of the mono-, di- or triphosphate esters of noribogaine, wherein the mono-, di- or triphosphate ester moiety is bonded to the 12-hydroxy group and/or the indole nitrogen of noribogame.
  • diphosphate refers to the group -P(0)(OH)-OP(0)(OH)2.
  • triphosphate refers to the group -P(0)(OH)- ( ⁇ ( ⁇ )( ⁇ )) 2 ⁇ .
  • impermissible substitution patterns e.g., methyl substituted with 5 fluoro groups or a hydroxy! group alpha to ethenylic or acetylenic unsaturation.
  • impermissible substitution patterns arc well known to the skilled artisan.
  • Such functional groups are well known in the art including acyl groups for hydroxy! and/or amino substitution, esters of mono-, di- and tri-phosphates wherein one or more of the pendent hydroxyl groups have been converted to an alkoxy, a substituted alkoxy, an aryloxy or a substituted aryloxy group, and the like.
  • “Pharmaceutically acceptable salt” refers to pharmaceutically acceptable salts, including partial salts, of a compound, which salts are derived from a variety of organic and inorganic counter ions well known in the art and include, by way of example only, sodium, potassium, calcium, magnesium, ammonium, tetraal ky 1 ammoni um , and the like; and when the molecule contains a basic functionality, salts of organic or inorganic acids, such as hydrochloride, hydrobromide.
  • tartrate mesylate, acetate, lactate, succinate, citrate, salicylate, malale, maleate, oxalate, phosphate, phosphite, sulfate, nitrate, perchlorate, aconitate, thalate, embonate, enanthate, and the like.
  • “Pharmaceutically acceptable partial salts” refers to compounds having a substituent capable of having more than one group form a salt but less than the maximum amount of such groups actually form a salt.
  • a diphospho group can form a plurality of salts and, if only partially ionized, the resulting group is sometimes referred to herein as a partial salt.
  • the term "therapeutically effective amount ' ' refers to the amount of a composition of this invention that is sufficient to effect treatment, as defined herein, when administered to a subject in need of such treatment.
  • the therapeutically effective amount will vary depending upon the subject and condition being treated, the weight and age of the subject, the severity of the condition, the particular composition or excipient chosen, the dosing regimen to be followed, timing of administration, the manner of administration and the like, all of which can be determined readily by one of ordinary skill in the art.
  • treatment means any treatment of a disease or condition in a patient, including:
  • the term "pain” refers to all types of pain, including neuropathic and nociceptive pain. It is also contemplated that the compositions disclosed herein can be used to treat other types of pain such as phantom pain which is the sensation of pain from a limb or organ that has been lost or from which a person no longer receives physical signals, and is an experience almost universally reported by amputees and quadriplegics.
  • addiction refers to a persistent behavioral pattern marked by physical and/or psychological dependency to a substance, particularly drugs such as narcotics, stimulants, and sedatives, including but not limited to heroin, ***e, alcohol, nicotine, caffeine, amphetamine, desoxyephedrine, methadone and combinations thereof.
  • treatment of addiction in a patient refers to reducing the withdrawal symptoms associated with drug dependency as well as alleviating drug cravings in addicts. Such symptoms include nausea, vomiting, anxiety, abdominal cramps, muscle pain, chills and headache.
  • the term "patient” refers to mammals and includes humans and non-human mammals.
  • This invention is relates to noribogaine derivative compounds. Such compounds are contemplated as being useful in treating pain and/or drug dependency. Accordingly, in one of its composition aspects, this invention is directed to a compound or a pharmaceutically acceptable salt thereof wherein said compound is represented by Formula IA or Formula IB, or a pharmaceutically acceptable salt of each thereof:
  • R is OR 1 or optionally substituted with 1 to 5 R 9 ;
  • R 1 is selected from the group consisting of hydrogen, -C(0)OX, -SO 2 OR 10 , a
  • X is C i -C alkyl optionally substituted with 1 to 5 R 9
  • each Y is independently selected from the group consisting of hydrogen, CVC 6 alkyl optionally substituted with 1 to 5 R 9 , C- 6 -Ci aryl optionally substituted w 7 ith 1 to 5 R J , C3-C 1 cycloalkyl optionally substituted with 1 to 5 R 9 .
  • Ci - 0 heteroaryl having 1 to 4 heteroatoms and which is optionally substituted with 1 to 5 R 9
  • Ci-C 10 heterocyclic having 1 to 4 heleroatoms and which is optionally substituted with 1 to 5 R 9
  • R' is halo, OR 20 , or Cj-C alkyl optionally substituted with 1 to 5 R 9 :
  • R 20 is selected from the group consisting of hydrogen , -C(0)X, -C(0)OX and -C(0)N(Y) 2 where X and Y are defined as above;
  • R 2 is hydrogen, -SO2OR 10 , a monophosphate, a diphosphate, or a triphosphate;
  • R 3 is selected from the group consisting of hydrogen, -(CH 2 ) m OR 7 , -CR 6 (OH)R 7 . -(CH 2 ) m CN, -(CH 2 ) m COR 7 , -(CH 2 ) m C0 2 R 7 , -(CH 2 ) m C(0)NR 6 R 7 , -( ( 1 l; ), ruleC ' ( ( ) )N ' N R R .
  • n 0, 1 , or 2;
  • L is a bond or C i -C 12 alkylene
  • R 4 is selected from the group consisting of C 1 -C 12 alkyl substituted with 1 to 5 R 9 , C 2 -Ci 2 alkenyl substituted with 1 to 5 R 9 , -X 2 -R 6 , -(X 2 -Y 2 ) n -X 2 -R 6 , -S0 2 NR 6 R 7 , -0-C(0)R 8 , -C(0)OR 7 , -C(0)NR 6 R 7 , -NR 6 R 7 ; -NHC(0)R 8 , and -NR 6 C(0)R 8 ;
  • X 2 is selected from the group consisting of O and S;
  • Y 2 is C 1 -C4 alkylene or C C 10 arylene, or a combination thereof;
  • n i , 2. or 3:
  • R b and R' are each independently selected from the group consisting of hydrogen.
  • -Ci2 alkyl optionally substituted with 1 to 5 R 9 , ( Cs heterocyclic having 1 to 4
  • R 8 is selected from the group consisting of C]-C 12 alkyl optionally substituted with 1 to 5 R 9 , Cs-Cehelerocycle having 1 to 4 heteroatoms optionally substituted with 1 to 5 R 9 , Cj- Ciocycloalkyl optionally substituted with 1 to 5 R 9 , C 6 -Ci aryl optionally substituted with 1 to 5 R 9 and C]-C 6 heteroaryl having 1 to 4 heteroatoms optionally substituted with 1 to 5 R 9 ;
  • R “ is selected from the group consisting of C1-C4 alkyl, phenyl, halo, -OH, -OR 10 , -CN, -COR 10 , -CO 2 R 10 ; -C(0)NHR 10 , -NR 10 R 10 , -C(O)NR ,0 R 10 . -C(0)NHNHR 10 .
  • R 10 is C1-C12 alkyl
  • R is -OH or C1-C12 alkyl optionally substituted with 1 to 5 R 9 , then R "5 is hydrogen;
  • the compound of Formula IA or IB is represented by Formula IA' and IB " respectively:
  • R, R R J and R 4 are as described for formula I.
  • R is halo, such as fluoro or chloro.
  • R is CrC 12 alkyl optionally substituted with 1 to 5 R 9 .
  • R is C1-C12 alkyl optionally substituted with a -S0 2 NR 6 R' group.
  • R is OR 1 ,
  • R 1 is selected from the group consisting of C(0)R s ; -C(0)NR 5 R :> and -C(0)QR 5 ; where each R 5 is independently selected from the group consisting of hydrogen, alkyl, alkenyl. alkynyl, aryl. heteroaryl, and hctcrocycle, and wherein the alkyl. alkenyl. alkynyl, aryl. heteroaryl, and heterocycle are optionally substituted with 1 to 5 R .
  • R 1 is -C(0)R ⁇ and wherein R 5 is Cj-Ci? alkyl or substituted C -C 12 alkyl or aryl. which C i to C alkyl or aryl is optionally substituted with C
  • R 5 is selected from the group consisting of methyl, ethyl, n-propyl, n-butyl, n-pentyl, n-hexyl, n-heptyl. n-octyl, n-nonyl, n-decyl. n-undecyl, n- dodecyl, iso-propyl sec -butyl, tcrt-butyl, phenyl, benzyl, methylbcnzyl, and methoxybenzyl.
  • R 1 is benzoyl
  • R 1 is hydrogen
  • R 2 is hydrogen. In some embodiments, R 2 is (> ( . * : alkyl optionally substituted with 1 to 5 R 9 , for example, CHiOH. In some embodiments, R 2 is aryl optionally substituted with 1 to 5 R 9 , for example, phenyl substituted with a halo group. In some embodiments, R 2 is -C(0)R ⁇ such as -C(0)CH 2 CH 2 N(CI l3)2. In some embodiments, R 2 is -C(0)NR 5 R 5 , such as -C(0)NHCH 3 or C(0)NHCH 2 CH:N(CH 3 )2. In some
  • R 2 is -C(0)OR 5 .
  • R 2 is -C(0)OR 5 .
  • -C(0)OCH 3 or -C(0)OCH 2 CH 2 N(CH 3 )2.
  • R is hydrogen or -CHbOH. In some embodiments, R is
  • L-R 4 is not ethyl. In some embodiments, L-R 1 is not alkyl. In some embodiments, when R is OR 1 , L-R 4 is not ethyl. In some embodiments, when R is OR 1 , L-R '1 is not alkyl.
  • L is Cs-CV,aIkyIene
  • R 4 is -X-R '
  • X is O
  • R 4 is selected from the group consisting of hydrogen, Ci -Cizalkyl, -CH 2 Ph, and -C(0)OR 7 .
  • R 4 is -NH 2 , -CILOCFf CHiOCLL or -OCH3.
  • J 066J the compound is selected from the group consisting of:
  • this invention is directed to a compound or a pharmaceutically acceptable salt thereof wherein said compound is represented by Formula 1IA or Formula IIB, or a pharmaceutically acceptable salt of each thereof:
  • R 11 is halo, -OH, -SH, -NH 2 , -S(0) 2 N(R 16 ) 2 , -X'-L'-R 17 , -X'-L'-R 18 , -X'-lJ-R 19 , or - X'-L'-CHR 17 R 18 , where X 1 is (>. S or NR 16 , or -O-Q, where Q is -S0 2 -(C,-Ci : alkyl). a monophosphate, a diphosphate, or a triphosphate;
  • L 1 is alkylene, arylene, -C(0)-alkylene, -C(0)-arylene, -C(0)0-arylene, -C(0)0- alkylene, -C(0)NR 19 -alkylene, -C(0)NR 19 -arylene, -C(NR l9 )NR ,9 -alkylene or
  • L 1 is configured such that -O-L'-R' ' is - OC(0)-alkylene-R 17 , -OC ⁇ 0)0-arylene-R N , -OC(0)0-alkylene-R 17 , -OC(O)- arylene-R 17 , -OC(0)NR I9 -alkylene-R 17 , -OC(0)NR l9 -arylene-R 17 , - OC( R L9 )NR l9 -alkylene-R 17 or -OC(NR 19 )NR l9 -arylene-R 17 , and wherein the alkylene and arylene are optionally substituted with 1 to 2 R 15 ;
  • R 12 is hydrogen, -S(()) : ( ) ". -S(0) 2 R 19 , -C(0)R 14 , -C(0)NR 14 R 14 , -C(0)OR 14 , C R C 12 alkyl optionally substituted with 1 to 5 R 15 , Ci-Ci 2 alkenyl optionally substituted with 1 to 5 R 15 , aryl optionally substituted with 1 to 5 R , -S0 2 - (Ci-Ci2 alkyl), a monophosphate, a diphosphate, or a triphosphate;
  • R 13 is hydrogen, halo, -OR 16 , -CN, ( . ⁇ -( ⁇ 2 alkyl, C]-Ci 2 alkoxy, aryl or aryloxy, where the alkyl, alkoxy, aryl, and aryloxy are optionally substituted with 1 to 5 R 15 ; each R 14 is independently selected from the group consisting of hydrogen, C1-C12 alkyl, C 2 -Ci 2 alkenyl, C 2 -Ci 2 alkynyl, aryl, heteroaryl, and heterocycle, and wherein the alkyl, alkenyl, alkynyl, aryl, heteroaryl, and heterocycle are optionally substituted with 1 to 5 R 13 ;
  • R 1" is selected from the group consisting of phenyl, halo, -OR 16 , -CN, -COR 15 ,
  • each R 16 is independently hydrogen or C 1 -C 12 alkyl optionally substituted with from 1 to 3 halo;
  • R 17 is hydrogen, -C(0)R 19 , ⁇ C(0)OR 19 , -C(0)N(R ,9 ) 2 or -N(R ,9 )C(0)R !9 ;
  • R 18 is hydrogen, -N(R I9 ) 2 , -C(0)N(R L9 ) 2 , -C(NR 19 )N(R I9 ) 2 , -C( S0 2 R 19 )N(R l9 ) 2 , -NR l9 C(0)N(R 19 ) 2 , -NR ,9 C(S)N(R ,9 ) 2 , -NR I9 C(NR ,9 )N(R I9 ) 2 ,
  • each R 19 is independently selected from the group consisting of hydrogen, C]-C
  • R i 2 and R 13 are hydrogen, then R 1 1 is not hydroxy:
  • R 13 when R 13 is hydrogen, R is -0-L ! -R , 7 5 -O-L'-R 1 8 , -O-L'-R 19 , and L 1 is
  • the compound of Formula II is represented by Formula IIC or Formula IID, or a pharmaceutically acceptable salt of each thereof:
  • R is halo. In certain embodiments of the compounds of Formula IIA, IIB, IIC, or IID. R 1 1 is bromo. In certain embodiments, R 1 1 is chloro. In certain embodiments of the compounds of Formula IIA, UB, IIC, or IID, R 1 1 is fluoro. In certain embodiments, R 1 1 is iodo.
  • R 1 ' is -OH, - O-L ' -R 1 7 , -O-L' -R 1 8 , -O-L' -R 19 or -0-L'-CHR 1 7 R , s .
  • R 1 1 is -SFI, -S-L'-R 1 ', -S-L'-R 18 , -S-L'-R 19 or -S-L'-CHR ⁇ R 18 .
  • R u is -NI 1 2 , -NR 16 -L !
  • R n is ⁇ S(0) 2 N(R 16 ) 2 .
  • 1 1 is -CI, - Br, -I, -OC(0)CH 2 CHj, -OC(0)CH 2 Ph, -OC(0)OCti 2 Cll 3 , -OC(0)OCI FPh,
  • R 12 is hydrogen
  • R 12 is - CH2CFI3, -CH2CHCH2, -CH 2 Ph, -C(0)0(CH 2 )2N(CH 3 )2 or -C(0)CH 2 (CH2)2S0 2 N(CH 3 )2.
  • R' is hydrogen
  • R 3 is halo. In certain embodiments of the compounds of Formula ILA, IIB, IIC, or IID, R 3 is bromo. In certain embodiments of the compounds of Formula IIA, IIB. IIC, or IID, R 3 is iluoro.
  • R' is -OCH 3 . -OCH2CH3. -OCFhPh or -CN.
  • the compound is represented by formula HE selected from Table 1 :
  • the compound is represented by Formula IC selected from Table 2:
  • the compound of Formula IA, IB, IC, IIA, IIB, IIC, or IID is the hydrochloride salt.
  • the compound of Formula IA, IB, IC, IIA, IIB, IIC, or IID is the hydrobromide salt.
  • the compound of Formula IA, IB, IC, IIA, IIB, IIC, or IID is the phosphate salt.
  • the compound of Formula IA, IB, IC, IIA, IIB, IIC, or IID is the sulfate salt.
  • the present invention is directed to a method for treating a pain in a patient which method comprises administering to said patient a compound of this invention or a pharmaceutically acceptable salt thereof or a pharmaceutical composition comprising a compound of this invention and a pharmaceutically acceptable excipient.
  • the pain can be any type of pain including, but not limited to neuropathic or nociceptive pain, and various types thereof including somatic, visceral and phantom pain.
  • the present invention is directed to a method for treating addiction in a patient which method comprises administering to said patient a compound of this invention or a pharmaceutically acceptable salt thereof or a composition comprising a compound of this invention and a pharmaceutically acceptable excipient.
  • the treatment of addiction in a patient comprises alleviating the symptoms associated with withdrawal from drug dependency.
  • symptoms include nausea, vomiting, anxiety, abdominal cramps, muscle pain, chills and headache.
  • treatment with a compound of this invention decreases the drug cravings normally experienced by addicts after cessation of the self administration of the abused substance.
  • the compositions disclosed herein are especially useful in the treatment of addiction to narcotics such as heroin and methadone. However, it is also useful in treating patients addicted to ***e, alcohol, amphetamines and combinations of these drugs.
  • the invention is also directed to a method for treating drug addiction (involving drug dependency or drug abuse) during withdrawal therapy by administering a compound of this invention to a patient at a dosage sufficient to reduce or eliminate one or more symptoms associated with withdrawal.
  • symptoms include nausea, vomiting, anxiety, abdominal cramps, muscle pain, chills and headache.
  • treatment with a compound of this invention is contemplated to decrease the drug cravings normally experienced by addicts after cessation of the self administration of the abused substance, for example, narcotics such as heroin and methadone.
  • compounds of this invention are contemplated to be also useful in treating patients addicted to ***e, alcohol, amphetamines and combinations of these drugs.
  • Compounds of this invention may be administered to patients suffering from drug dependence or abuse in conjunction with an opioid antagonist such as naloxone, naltrexone or nalorphine, for example, at a concentration of between 0.1 5 mg and 0.5 mg for each mg of the compound of this invention administered.
  • an opioid antagonist such as naloxone, naltrexone or nalorphine
  • Tobacco use remains a major health problem despite widespread knowledge of the damaging consequences. Although current smoking cessation therapies, including nicotine replacement, bupropion, and varenicline, have had some success, they are inadequate since only a fraction (30-40%) of smokers who try these medications abstain from tobacco use. Consequently, more effective or add-on therapies are needed. Nicotine is one of the most addictive drugs that is widely used; 95% or more of its users with a strong desire to stop using relapse within one year. Chronic nicotine use leads to physiological changes in nAChR function and expression, including upregulation of high-affinity a4b2 nAChRs expression and reduced receptor function due to desensitization.
  • the physical discomfort associated with nicotine withdrawal includes severe cravings, anxiety, dysphoria, and autonomic dysfunction.
  • the reinforcing and addictive eff ects of nicotine are due to is actions on nAChRs, the
  • nAChR subtypes play a role in the various aspects of nicotine dependence/withdrawal, and their validation as drug targets for medication development remains a complex area of investigation.
  • the a4 subunit is implicated in nicotine reinforcement, sensitization and the development of tolerance. While the development of tolerance is not regulated by a7 nAChRs, a recent stud indicated that that these receptors may control the severity of the withdrawal symptoms. Thus, there is a complex regulatory interplay of nAChRs, which likely contribute to nicotine dependence/withdrawal.
  • nAChRs Neuronal nicotinic acetylcholine receptors
  • Varenicline is a weak partial agonist for a4b2 nAChR subtypes and a less potent agonist at a7 subtypes.
  • a serendipitous finding in aiding smoking cessation is the discovery that the atypical anti-depressant bupropion, whose mechanism of action as a non-competitive antagonist of a3b4 nAChRs and antidepressant activity underlie its efficacy.
  • Buproprion analogs are under development as potential pharmacotherapies for smoking cessation.
  • Second generation noribogaine analogs targeted to a3b4 nAChRs offer a novel synthetic route for development of potential small molecules to treat tobacco addiction and other SRDs.
  • SNPs in the gene cluster CHRNA5/A3/B4, encoding for the a3, a5 and b4 nAChR subunits are associated with increased risk for heavy smoking, inability to quit, and increased sensitivity to nicotine. Furthermore, a3b4 nAChRs or other b4 containing receptors have been suggested to be involved in nicotine withdrawal.
  • the expression of the a3b4 AChR is restricted to a few discrete brain areas, including the medial habenula and interpeduncular nucleus, and autonomic ganglia.
  • the present invention is directed to a method of treating nicotine addiction in a patient which method comprises administering an effective amount of a compound of this invention or a pharmaceutically acceptable salt thereof or a composition comprising a compound of this invention and a pharmaceutically acceptable excipient to a patient in need thereof.
  • the present invention is directed to a method of inhibiting a3b4 nicotinic acetylcholine receptors which method comprises administering an effective amount of a compound of this invention or a pharmaceutically acceptable salt thereof or a composition comprising a compound of this invention and a pharmaceutically acceptable excipient.
  • Compounds of this invention maybe used alone or in combination with other compounds to treat pain and/or addiction.
  • the coadministration can be in any manner in which the pharmacological effects of both are manifest in the patient at the same time.
  • co-administration does not require that a single pharmaceutical composition, the same dosage form, or even the same route of administration be used for administration of both the compound of this invention and the other agent or that the two agents be administered at precisely the same time.
  • coadministration will be accomplished most conveniently by the same dosage form and the same route of administration, at substantially the same time. Obviously, such administration most advantageously proceeds by delivering both active ingredients simultaneously in a novel pharmaceutical composition in accordance with the present invention.
  • a compound of this invention can be used as an adjunct to conventional drug withdrawal therapy, specifically providing for the administration of a compound of this invention with one or more opioid antagonists.
  • compositions comprising a pharmaceutically acceptable excipient and a compound of this invention or mixtures of one or more of such compounds.
  • compositions suitable for oral, intravenous or intraarterial delivery will probably be used most frequently, other routes that may be used include peroral, pulmonary, rectal, nasal, vaginal, lingual, intramuscular, intraperitoneal, intracutaneous and subcutaneous routes.
  • the composition can be administered transdermally in which drug is applied as part of a cream, gel, or patch (for examples of transdermal formulations, see U.S. Pat. Nos. 4,806,341 ; 5, 149,538; and 4,626,539).
  • dosage forms include tablets, capsules, pills, powders, aerosols, suppositories, parenterals, and oral liquids, including suspensions, solutions and emulsions. Sustained release dosage forms may also be used, for example, in a transdermal patch form. All dosage forms may be prepared using methods that are standard in the art (see e.g., Remington's Pharmaceutical Sciences, 16th ed., A. Oslo editor, Easton Pa. 1980). Intranasal administration is an effective method for delivering a therapeutic agent directly to the respiratory tract, where the therapeutic agent may be quickly absorbed.
  • compositions are comprised of in general, a compound of this invention or a mixture thereof in combination with at least one pharmaceutically acceptable excipicnt.
  • Acceptable excipients are non-toxic, aid administration, and do not adversely affect the therapeutic benefit of the compound of this invention.
  • excipients may be any solid, liquid, semi-solid or, in the case of an aerosol composition, gaseous excipient that is generally available to one of skill in the arl.
  • Pharmaceutical compositions in accordance with the invention are prepared by conventional means using methods known in the art.
  • Solid pharmaceutical excipients include starch, cellulose, talc, glucose, lactose, sucrose, gelatin, malt, rice, flour, chalk, silica gel, magnesium stearate, sodium stearate, glycerol monoslearate, sodium chloride, dried skim milk and the like.
  • Liquid and semisolid excipients may be selected from glycerol, propylene glycol, water, ethanol and various oils, including those of petroleum, animal, vegetable or synthetic origin, e.g., peanut oil, soybean oil, mineral oil, sesame oil, etc.
  • Preferred liquid carriers, particularly for injectable solutions include water, saline, aqueous dextrose, and glycols.
  • Compressed gases may be used to disperse a compound of this invention in aerosol form.
  • Inert gases suitable for this purpose are nitrogen, carbon dioxide, etc.
  • Other suitable pharmaceutical excipients and their formulations are described in Remington's
  • compositions disclosed herein may be used in conjunction with any of the vehicles and cxcipicnts commonly employed in pharmaceutical preparations, e.g., tale, gum arable, lactose, starch, magnesium stearate, cocoa butter, aqueous or non-aqueous solvents, oils, paraffin derivatives, glycols, etc. Coloring and flavoring agents may also be added to preparations, particularly to those for oral administration. Solutions can be prepared using water or physiologically compatible organic solvents such as ethanol, 1 ,2-propylene glycol, polyglycols, dimethylsulfoxide. fatty alcohols, triglycerides, partial esters of glycerine and the like.
  • Parenteral compositions containing noribogaine may be prepared using conventional techniques that may include sterile isotonic saline, water, 1 ,3-butanediol, ethanol, 1 ,2- propylene glycol, polyglycols mixed with water, Ringer's solution, etc.
  • the amount of the compound in a formulation can vary within the full range employed by those skilled in the art.
  • the formulation will contain, on a weight percent (wt %) basis, from about 0.01 to 99.99 wt % of a compound of this invention based on the total formulation, with the balance being one or more suitable pharmaceutical excipients.
  • the compound is present at a level of about 1 to 80 wt %.
  • a compound of this invention should generally be present in such compositions at a concentration of between about 0.1 and 20 mg/ml.
  • naloxone or naltrexone When either naloxone or naltrexone is combined with a compound of this invention, they should be present at 0.05 to 0.5 mg for each mg of the compound of this invention.
  • the choice of formulation depends on various factors such as the mode of drug administration and bioavailability of the drug substance.
  • the compound can be formulated as liquid solution, suspensions, aerosol propellants or dry powder and loaded into a suitable dispenser for administration.
  • suitable dispenser for administration There are several types of pharmaceutical inhalation devices-nebulizer inhalers, metered dose inhalers (MDI) and dry powder inhalers (DPI).
  • MDI metered dose inhalers
  • DPI dry powder inhalers
  • Nebulizer devices produce a stream of high velocity air that causes the therapeutic agents (which are formulated in a liquid form) to spray as a mist that is carried into the patient's respiratory tract.
  • MDI's typically are formulation packaged with a compressed gas. Upon actuation, the device discharges a measured amount of therapeutic agent by compressed gas.
  • DPI dispenses therapeutic agents in the form of a free flowing powder that can be dispersed in the patient's inspiratory air-stream during breathing by the device.
  • the therapeutic agent is formulated with an excipient such as lactose.
  • a measured amount of the therapeutic agent is stored in a capsule form and is dispensed with each actuation.
  • U.S. Patent No. 5,145,684 describes the production of a pharmaceutical formulation in which the drug substance is pulverized to nanopartieles (average particle size of 400 nm) in the presence of a surface modifier and then dispersed in a liquid medium to give a pharmaceutical formulation that exhibits remarkably high bioavailability.
  • the compound of this invention is administered in an effective amount. It is contemplated that the dosage required for treating pai n or addition, or a combination thereof may differ according to the condition(s) being treated, however, the dosing regimen can be readily determined by the attending clinician based on the desired treatment. It is contemplated that generally, the dosage of a compound of this invention administered to a patient may be from about 0.01 to about 1000 mg per kg of body weight per day (mg/kg/day), or from 0.05 to 500 mg/kg/day, preferably, from about 0.1 to about 100 mg/kg/day, more preferably from about 0.5 to 50 mg/kg/day. For example, for administration to a 70 kg person, the dosage range would preferably be about 35 to 700 mg per day.
  • the present invention is directed to a pharmaceutical composition, preferably in unit dose form, comprising a compound of this invention.
  • a pharmaceutical composition preferably in unit dose form, comprising a compound of this invention.
  • one or more unit doses provide an amount of a compound of this invention effective to treat pain and/or addition.
  • composition administered will depend on a number of factors, including but not limited to the desired final concentration of the compound, the
  • the active compound is effective over a wide dosage range and is generally administered in a pharmaceutically effective amount. It will be understood, however, that the amount of the compound actually administered will be determined by a physician, in the light of the relevant circumstances, including the condition to be treated, the chosen route of administration, the actual compound administered, the age, weight, and response of the indi vidual patient, the severity of the patient's symptoms, and the like.
  • compositions are administered in one dosing of a single formulation and in other embodiments, compositions are administered in multiple dosing of a single formulation within a specified time period.
  • the time period is between about 3 hours to about 6 hours. In other embodiments, the time period is between about 6 hours and 12 hours. In additional embodiments, the time period is between about 12 hours and 24 hours. In yet further embodiments, the time period is between about 24 hours and 48 hours.
  • the administration of separate formulations can be simultaneous or staged throughout a specified time period, such that all ingredients are administered within the specified time period.
  • Example 1 Tablet formulation
  • a suppository of total weight 2.5 g is prepared by mixing the compound invention with Witepsol® H- 15 (triglycerides of saturated vegetable fatty acid;
  • Example 6 Screening compounds for nAChR target
  • the FIJPR® Ca+2 flux assay is used with IIEK293 cells over- expressing a3b4, a4b2 or a7 to determine the potency and selectivity of the noribogaine congeners.
  • the HEK293 (a3b4, a4b2 and a7) and SH-SY5Y cell lines (a3b4) expressing nAChRs are used.
  • Validated assays optimized with respect to sensitivity, dynamic range, signal intensity and stability for chemical probe validation and SAR refinement are used.
  • Nicotinic functional assays are performed using the Molecular Devices FLIPR® Calcium 5 Assay Kit. HEK293 cells overexpressing ⁇ 3 ⁇ 4, ⁇ .4 ⁇ 2 or u7 (50,000 cells per well in 100 ⁇ growth media) and are plated in clear bottom black walled 96-well microplates and incubated for 24-48 hours at 37°C, 5% C0 2 until a 80-90% confluent monolayer is formed. On the day of the assay, 100 ⁇ of Calcium 5 Dye is added to each well. Following addition of dye. cells are incubated at 25°C for 60 minutes.
  • 86 RB + efflux assays are also used to assess whether noribogaine or its analogs have activity as antagonists at human nAChR using naturally or heterologously expressed cell lines and compared to synaptosomal preparations from mouse brain. SHSY cells function only for measures of a3b4, because a7 nACliRs, which are also expressed by these cells inactivate too quickly to detectably contribute to ion influx. 86 RB + efflux in the presence of the compounds is used to test for agonist and antagonist activity at hAChRs. Representative concentration curves are obtained to complete the SAR profiling of the compound series.
  • Inhibition of agonist stimulated DA and ACh release are tested in striatal and IPN synaptosomal preparations according to known methods using L-nicotine or cy tosine to stimulate release, respectively.
  • IC50 values for inhibition of ACh and DA release ( ⁇ ) are determined from the curve fits of the data with or without test compound present.
  • Example 7 nui and kappa opioid receptor binding activity
  • Noribogaine binds with high affinity (50 n ) to the 5-HT transporter and weak affinity at mu (900 nM) and kappa (1 ⁇ ) opioid receptors.
  • noribogaine only bound to SERT, DAT, mu and kappa opioid receptors (out of a panel of over 50 receptors, transporters, ion channels, second messengers, growth factor receptors and enzymes).
  • SERT, DAT, mu and kappa opioid receptors out of a panel of over 50 receptors, transporters, ion channels, second messengers, growth factor receptors and enzymes.
  • Noribogainc analogs will likely retain affinity for the 5-HT transporter and we plan to fully characterize their activity in radioligand binding and functional uptake assays (serotonin and DA).
  • Radiolabeling of opioid receptors, and biogenic amine transporters utilizes conventional radioligands, buffers and incubation conditions according to published assays.
  • Test compounds, reference compounds and the radioligand are added to binding buffer. Assays are run with test or reference compound over a range of concentration (0.0001 -10 uM) in triplicate using a 96-well sample plate containing radioligands specific for the target receptor or transporter. Cell membranes at the appropriate concentration are added in a volume of 400 ⁇ in binding buffer. Following mixing on a plate shaker, samples are incubated for 1 -2 hours (depending on the transporter or receptor) at 25°C and filtered onto 96-well Unifilters (pre-treated with 0.3% polyefhyleneimine) using a Filtermate harvester. Bound radioactivity is counted in a Microbeta scintillation counter.
  • Total bound radioactivity is estimated from triplicate wells containing no test or reference compound, together with nonspecific binding and filter blanks.
  • the (( ' ;,, and i values for test and reference compounds are obtained by fitting normalized CPM data fitted using nonlinear (Graphpad Prism software).
  • Example 8 CNS penetration across the brain blood barrier
  • the compounds of this invention can be prepared from readily available starting materials using the following general methods and procedures. It will be appreciated that where typical or preferred process conditions (i.e., reaction temperatures, times, mole ratios of reactants, solvents, pressures, etc.) are given, other process conditions can also be used unless otherwise stated. Optimum reaction conditions may vary with the particular reactants or solvent used, but such conditions can be determined by one skilled in the art by routine optimi ation procedures.
  • the compounds of this invention will typically contain one or more chiral centers. Accordingly, if desired, such compounds can be prepared or isolated as pure stereoisomers, i.e., as individual enantiomers or diastereomers, or as stereoisomer-enriched mixtures. All such stereoisomers (and enriched mixtures) are included within the scope of this invention, unless otherwise indicated. Pure stereoisomers (or enriched mixtures) may be prepared using, for example, optically active starting materials or stereoselective reagents well-known in the art. Alternatively, racemic mixtures of such compounds can be separated using, for example, chiral column chromatography, chiral resolving agents and the like.
  • Scheme 1 shows an exemplary general process for prepare compounds of this invention.
  • the starting materials for the following reactions are generally known compounds or can be prepared by known procedures.
  • Scheme 1 shows an exemplifying procedure to prepare compounds of Formula I, wherein R 2 , R 3 , L and R 4 are as defined herein.
  • Compound 1.1 wherein Pr 1 is R 1 as defined herein or a hydroxy protecting group, and Pr' is R as defined herein or a protecting group for the indole nitrogen, is converted to Compound 1.2 using the procedure described in uehne, et al. J. Org. Chem. 50:919 (1985).
  • Hydroxyl protecting groups include a hydrolysable group as defined for Formula I, such as -C(0)R.
  • R is as defined herein, and other hydroxyl protecting groups known in the art, such as a benzyl (Bn) group, 2,4-dichlorobenzyl (DCB), methoxymethyl (MOM), tetrahydropyranyl (THP), acyl (such as acetyl (Ac)) or a silyl group, etc.
  • hydroxyl protecting groups such as a benzyl (Bn) group, 2,4-dichlorobenzyl (DCB), methoxymethyl (MOM), tetrahydropyranyl (THP), acyl (such as acetyl (Ac)) or a silyl group, etc.
  • Suitable hydroxy protecting groups are known in the art.
  • Protecting groups for the indole nitrogen include arylsulfonyl derivatives (e.g. tosyl (Ts)), carbamates (e.g.
  • fluorenylmethyloxycarbonyl Fmoc
  • t-BOC tert-butyloxycarbonyl
  • trialkylsilyl groups e.g. triisopropylsilyl
  • ⁇ , ⁇ -acetals e.g. (trimethylsilyl)ethoxymethyl ether (SEM)
  • certain substituted alkyl groups e.g. benzyl
  • Pr 1 and/or Pr 2 When Pr 1 is R 1 and/or Pr 2 is R 2 , they can be retained as part of the final compounds, or may be converted to other groups as defined in Formula I by methods generally known in the art. When Pr 1 and/or Pr 2 are other protecting groups, they can be removed under proper deprotection conditions to provide the free hydroxy group and/or free indole nitrogen, which may be further converted to the R 1 and/or R 2 groups as defined in Formula I.
  • Compound 1.2 may react with Compound 1.3 to give Compound 1.4 in dry toluene under reflux conditions, and preferably with a Dean-Stark trap with molecular sieves.
  • Compound 1.3 can be prepared by using methods described in U.S. Patent 6,21 1 ,360, which is incorporated by reference in its entirety.
  • Compound 1.2 may react with Compound 1.3 to give a
  • condensation product in an organic solvent such as alcohol solvents, such as methanol, ethanol, isopropanol, and n-butanol; ester-containing solvents, such as ethyl acetate and isopropyl acetate; ether solvents, such as tetrahydrofuran, diglyme, and dioxane; chlorinate hydrocarbons, such as methylene chloride, chloroform, and carbon tetrachloride; aromatic hydrocarbons, such as benzene, toluene, and xylene; acetonitrile: pyridine; and
  • the reaction may proceed under room temperature or under heated conditions, such refiexing conditions.
  • the condensation product is treated in a suitable solvent with an equivalent amount of an appropriate arylalkyl containing a good leaving group, such as an arylalkyl tosylate, an arylalkyl mesylate, or an arylalkyl halide, such as benzyl bromide, for 0.5 to 72 hours, for example, 16 hours, at 50 L C to 120 °C, which may be at the reflux temperature of the solvent.
  • Suitable solvents include lower alkanes, such as pentane.
  • hcxanc or petroleum ether: aromatic hydrocarbon solvents, such as benzene, toluene, and xylene; alcohols, such as methanol, ethanol, isopropanol, and n-butanol; and ether solvents, such as diethyl ether, diglyme, or tetrahydrofuran.
  • aromatic hydrocarbon solvents such as benzene, toluene, and xylene
  • alcohols such as methanol, ethanol, isopropanol, and n-butanol
  • ether solvents such as diethyl ether, diglyme, or tetrahydrofuran.
  • This reaction can proceed in an organic solvent, including alcohol solvents, such as methanol, ethanol, isopropanol, and n-butanol; ketone solvents, such as acetone, methyl ethyl ketone, and cyclopentanone; ester-containing solvents, such as ethyl acetate and isopropyl acetate; ether solvents, such as tetrahydrofuran, diglyme, and dioxanc; chlorinated hydrocarbons, such as methylene chloride, chloroform, and carbon tetrachloride; acetonitrile; pyridine; and dimethylformamide.
  • alcohol solvents such as methanol, ethanol, isopropanol, and n-butanol
  • ketone solvents such as acetone, methyl ethyl ketone, and cyclopentanone
  • ester-containing solvents such as ethyl acetate and isopropyl
  • This reaction can be conducted at any temperature from room temperature to the boiling point of the solvent, for example, from 50 °C to 70 U C for, for example, from 1 to 10 hours.
  • the transient enamine acryiate spontaneously cyclizes to give Compound 1.4.
  • Compound 1.4 undergoes reduction with a reducing agent, such as NaBH 4 , in the presence of acetic acid, under heating conditions, for example, at a temperature of between 80 °C and 1 10 °C, or between 85 °C and 95 °C, followed by catalytic hydrogenation in the presence of a catalyst, such as palladium, in a solvent, such as methanol, to provide Compound 1.5.
  • a reducing agent such as NaBH 4
  • acetic acid for example, at a temperature of between 80 °C and 1 10 °C, or between 85 °C and 95 °C
  • a catalyst such as palladium
  • Pr 1 and/or Pf may be removed under suitable deprotection conditions to provide compounds where R 1 and/or are hydrogen.
  • R 1 and/or are hydrogen.
  • One or both of these hydrogen atoms may be substituted by a desired R 1 and/or R 2 by reaction with R'Lg and/or R 2 Lg, wherein Lg is a leaving group, such as a halo group (e.g., chloro, bromo and iodo), sulfonate esters, such as para-toluenesulfonate or tosylate (OTs), hydroxy or alkoxy group.
  • a halo group e.g., chloro, bromo and iodo
  • sulfonate esters such as para-toluenesulfonate or tosylate (OTs)
  • OTs para-toluenesulfonate
  • OTs para-toluenesulfonate
  • Compound 2.2 wherein Pr l and Pr 2 are as defined above.
  • Compound 2.2 is reduced to the corresponding alcohol Compound 2.3, using a reducing agent, such as lithium
  • Reduction of Compound 2.2 to Compound 2.3 may optionally conducted via an intermediate aldehyde compound.
  • the hydroxy group of Compound 2.3 is converted to a leaving group Lg 2 , such as a halo group (e.g., chloro. bromo and iodo), sulfonate esters, such as para-toluenesulfonate or tosylate (OTs), to give Compound 2.4.
  • Compound 2.4 can react with ammonium to give Compound 1.1 or with a protected ammonium compound, such as NH(t-Boc)2 to give Compound 1.1 after deprotection of the protecting group.
  • a protected ammonium compound such as NH(t-Boc)2
  • Other known methods of converting the acid group (-C(O)OH) to an methyl amino group (-CH 2 NH 2 ) can also be used.
  • the bromo compound corresponding to Compound 1.1 can be prepared, which can be used to replace Compound 1.1 in Scheme 1 to prepare compounds of this invention wherein R is bromo.
  • the bromo atom can be converted to other R groups as defined for f ormula 1 using procedures known in the art.
  • reaction with a suitable agent such as haloalkyk haloalkenyl, or haloalkynyl, optionally in the presence of a copper (I) or palladium catalyst and/or a base, such as triethylamine, would give compounds where R is alkyl (when using haloalkenyl or haloalkynyl, the double bond or triple bond can be reduced by hydrogcnation).
  • Scheme 3 shows an example of preparing an intermediate Compound 3.5, which may be used in Scheme 1 to replace Compound 1.1 to prepare compounds of this invention where R is chloro.
  • Compound 3.1 (5-chloroindole-3-carboxaldehyde, which is commercially available, for example, from Sigma- Aldrich Corp.) is protected with Pr 2 to give Compound 3.2.
  • Compound 3.2 may react with a Wittig agent, e.g. PhsPCff ⁇ B . to give Compound 3.3.
  • Compound 3.3 is converted to Compound 3.4, wherein Halo is a halo such as Br or CI. by reacting with, for example, hydrogen bromide (HBr).
  • HBr hydrogen bromide
  • Compound 3.4 can react with ammonium to give Compound 3.5 or with a protected ammonium compound, such as NH(t-Boc) 2 to give Compound 3.5 after deprotection of the protecting group.
  • a protected ammonium compound such as NH(t-Boc) 2
  • Other known methods of converting the acid group (-C(O)OH) to an methyl amino group (- CH 2 NH 2 ) can also be used.
  • LG is a leaving group such as hydroxy, alkoxy, halo, etc., to give Compound 4.2, which may further react with LG-R 12 to form Compound 4.3,
  • the C12-phenol, thiol, or amino group is protected by reaction with a suitable protecting group, PG-LG. where LG is a leaving group such as defined above, such that the indole nitrogen is selectively derivatized with R 12 .
  • suitable protecting groups are well kno B in the art (see T. W. Greene, P. G. M.
  • certain compounds of this invention may be prepared by reacting ibogaine with LG-R 12 to give Compound 5.1.
  • Compound 5.1 can be demethylated by methods known in the art, such as reaction with boron tribromide/methylene chloride at room temperature to give Compound 5,2, which may further react with LG ⁇ R ⁇ '° to give Compound 5.3, Scheme 5
  • the carboxymethyl group in Voacangine is hydrolyzed or converted into litium salt using an organolithium reagent such as butyllithium and proapancthiol in a solvent such as HMl'A/ I HF.
  • the lithium salt is then converted to the acid chloride 6,2 by reacting with oxalyl chloride in presence of a base such as pyridine in a solvent such as THF.
  • the acid chloride 6.2 is then esterfied with an alcohol to give the ester 6.3.
  • Demethylalion of the ester with a reagent such as BBr;, in a solvent such as methylene chloride gives compounds of formula IC.
  • certain compounds of this invention may be prepared using noribogaine, which may be prepared according to known procedures, such as by demethylating ibogaine by methods known in the art, such as reaction with boron tribromi de/methy 1 ene chloride at room temperature.
  • Schemes 7 and 8 below show reaction schemes for the sulfation of the 12 -hydroxy 1 group and optionally for the disulfation of the 12-hydroxyl group and the indole nitrogen atom.
  • Scheme 8 below shows reaction schemes for selective sulfation of the indole nitrogen atom by protecting the 12-hydroxyl group with a conventional hydroxyl protecting group.
  • a variety of protecting groups, preferably those stable under acidic conditions are useful as the Pg, as will be apparent to the skilled artisan.
  • An ester of the chlorosulfonic acid may be used to prepare an ester of the compound of Formula I.
  • indole nitrogen of noribogaine can be protected, the sulfation carried out on the hydroxy group of noribogaine, following which, the N-protecting group is deprotected.
  • Methods for preparing the N-protected noribogaine will be apparent to the skilled artisan in view of this disclosure.
  • X refers to a leaving group such a chloro, bromo, iodo, or a R 5 - S0 3 -moiety, where R s ia Ci-C 6 alkyl optionally sublimed with 1 -3 fluoro atoms or R s is phenyl optionally substituted with 1-3 halo or Ci-C 6 alkyl groups.
  • the dihydronoribogaine compounds of Formula IB are synthesized by reducing the double bond of the corresponding noribogaine derivative.
  • Various reducing agents well known to the skilled artisan are useful for this purpose.
  • cataly tic hydrogenalion employing hydrogen and a catalyst such as Pd/C or Pl/C is useful for providing the 9, 17 as. i.e. the a,a or the ⁇ , ⁇ dihydro compounds.
  • Reagents such as borohydride or aluminum hydrides are useful for providing the ⁇ , ⁇ or the ⁇ , ⁇ dihydro compounds.
  • Compounds of this invention as represented by Formula 1 -1 can be prepared from noribogaine using an appropriate phosphate source, such as phosphoric acid or a
  • phosphoramidite such as di-fcr/-butyl N,N-diisopropylphosphoramidite.
  • Compounds of Formula 1-2 can be prepared from compounds of Formula 1-1 using an appropriate phosphate source under known reaction conditions. The reactions are carried out for a period of time sufficient to provide a substantial amount of the product, which can be ascertained by- using routine methods such as thin layer chromatography, ⁇ -nuclear magnetic resonance (NMR) spectroscopy, and the likes.
  • Compounds of Formula 1-1 and 1-2 can be isolated and optionally purified using standard purification techniques, such as liquid chromatography.
  • Scheme 2 follows much of the chemistry of Scheme 1 with the exception that a blocking (protecting group - Pg) is used to avoid phosphorylation of the 12 hydroxyl group.
  • the dihydronoribogaine compounds of Formulas 1, 1-A, and 11 are synthesized by reducing the corresponding double bond of noribogaine.
  • Various reducing agents well known to the skilled artisan are useful for this purpose.
  • catalytic hydrogenalion employing hydrogen and a catalyst such as Pd/C or Pt/C is useful for providing the 9, 1 cis, i.e. the ⁇ , ⁇ or the ⁇ , ⁇ dihydro compounds.
  • Reagents such as borohydride or aluminum hydrides are useful for providing the ⁇ , ⁇ or the ⁇ , ⁇ dihydro compounds.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Medicinal Chemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Epidemiology (AREA)
  • Addiction (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Biomedical Technology (AREA)
  • Neurology (AREA)
  • Neurosurgery (AREA)
  • Psychiatry (AREA)
  • Emergency Medicine (AREA)
  • Toxicology (AREA)
  • Pain & Pain Management (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Nitrogen Condensed Heterocyclic Rings (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

This invention relates noribogaine derivatives, compositions and methods of use thereof.

Description

SUBSTITUTED NORIBOGAINE
Field of the Invention
[0001 ] This invention relates noribogaine derivatives, compositions and methods of use thereof.
State of the Art
|0002| Noribogaine is a well known derivative of ibogaine and is sometimes referred to as 12-hydiOxyibogaine. It is a metabolite of ibogaine. U.S. Patent No. 2,813,873 claims noribogaine albeit as 12-O-deinethylibogaine" while providing an incorrect structural formula for ibogaine. The structure of noribogaine has now been thoroughly evaluated and is found to combine the features of tryptamine, tetrahydrohavaine and indolazepines.
Noribogaine can be depicted by the following formula:
Figure imgf000002_0001
[0003] Noribogaine and its pharmaceutically acceptable salts have recently received significant attention as a non-addictive alkaloid useful in treating addiction (U.S. Patent No. 6.348,456) and as a potent analgesic (U.S. Patent No. 7,220,737).
[0004] Noribogaine has been found to have properties suitable for the treatment of pain and to the withdrawal symptoms associated with drug dependency or abuse. In particular, it is believed that noribogaine binds to two classes of opioid receptors that have been associated with pain relief, the μ and κ receptors. In the case of the μ-type receptors, it appears that noribogaine acts as an opiate agonist. In addition, noribogaine elevates brain serotonin levels by blocking synaptic reuptake. It is believed that such levels (as well as ligand interactions at the μ and κ opiate receptors) play a role in the anxiety and drug cravings experienced by addicts during withdrawal,
[00051 Noribogaine analogs are also found to inhibit a3b4 nicotinic acetylcholine receptors (nAChRs).
[0006] Noribogaine analogs are also found effective for treatment of nicotine addiction and for treatment of other substance abuse related disorders (SRDs). Summary of the Invention
[0007) This invention relates to noribogaine derivative compounds. Such compounds are contemplated as being useful in treating drug addiction and/or pain. Accordingly, in one of its compound aspects, this invention is directed to a compound or a pharmaceutically acceptable salt thereof wherein said compound is represented by Formula IA or Formula IB, or a pharmaceutically acceptable salt of each thereof:
Figure imgf000003_0001
IA IB
wherein
R is OR or Ci-C]2 alkyl optionally substituted with 1 to 5 R ;
R1 is selected from the group consisting of hydrogen, -C(0)OX, -S02OR , 1'0. a
monophosphate, a diphosphate, a triphosphate, and -C(0)N(Y)2 where X is Ci -C6 alkyl optionally substituted with 1 to 5 R9, and each Y is independently selected from the group consisting of hydrogen, C)-C6 alkyl optionally substituted with 1 to 5 R9, C6-C] 4 aryl optionally substituted with 1 to 5 R9, C3-C10 cycloalkyl optionally substituted with 1 to 5 R9, Cj-Ci o heteroaryl having 1 to 4 heteroatoms and which is optionally substituted with 1 to 5 R9, d-C 10 heterocyclic having 1 to 4 heteroatoms and which is optionally substituted with 1 to 5 R9. or where each Y. together with the nitrogen atom bound thereto form either a C1 -C6 heterocyclic having 1 to 4 heteroatoms and which is optional ly substituted with 1 to 5 R9, or a Ci-Cg heteroaryl having 1 to 4 heteroatoms and which is optionally substituted with 1 to 5 R9;
R' is halo. OR20, or Ci-C 12 alkyl optionally substituted with 1 to 5 R9;
R2 is selected from the group consisting of hydrogen . -C(0)X, -C(0)OX and -C(0)N Y )2 where X and Y are defined as above;
R2 is hydrogen, -SO2ORi . a monophosphate, a diphosphate, or a triphosphate;
RJ is selected from the group consisting of hydrogen, -(CFl2)mOR ', -CR6(()H)R7, -(CH2)mCN, -(CH2)mCOR7, -(CH2)mC02R7, -(CH2)mC(0)NR6R7, ~(CFI2)mC(0)NR6NR7R7, m is 0, 1 , or 2;
L is a bond or C 1 -C 12 alkylene;
R4 is selected from the group consisting of C1-C12 alkyl substituted with 1 to 5 R9, C2-C12 alkenyl substituted with 1 to 5 R9, -X2-R6, -(X2-Y2)n-X2-R6, -S02NR6R7, -0-C(0)R8, -C(0)OR7, -C(0)NR6R7, -NR6R\ -NHC(0)R8, and -NR6C(0)R8;
X2 is selected from the group consisting of O and S;
Y2 is C 1-C4 alkylene or QJ-C IQ arylene, or a combination thereof;
n is 1, 2, or 3;
R6 and R' are each independently selected from the group consisting of hydrogen, C1-C12 alkyl optionally substituted with 1 to 5 R9, C1 -C6 heterocycle having 1 to 4 heteroatoms which is optionally substituted with 1 to 5 R9, C3-Cio cycioalkyl optionally substituted with 1 to 5 R9, Ce-Cio aryl optionally substituted with 1 to 5 R9 and Ci- heteroaryl having 1 to 4 heteroatoms optionally substituted with 1 to 5 R9; or
R° and R' are joined to form (.>(.'„ heterocycle having 1 to 4 heteroatoms which is optionally- substituted with 1 to 5 R9:
R is selected from the group consisting of C|-C 12 alkyl optionally substituted with 1 to 5 R , C1-C heterocycle having 1 to 4 heteroatoms optionally substituted with 1 to 5 R9, C3- Cio cycloalkyl optionally substituted with 1 to 5 R9, Ce-Cjo aryl optionally substituted with 1 to 5 R9 and Ci-Ce heteroaryl having 1 to 4 heteroatoms optionally substituted with 1 to 5 R9:
R9 is selected from the group consisting of C1 -C4 alkyl, phenyl, halo, -OH, -OR10, -CN,
-COR10, -C02R10, -C(0)NHR10, -NR10R10, -CCOJNR^R10, -C(0)NHNHR10,
-CCOJNR^NHR10, -C(O)NR10NR,0R10, -C(O)NHNR,0C(O)R10,
-C(0)NHNHC(0)R10, -SO2NR10R10, -C(O)NR,0NRi0C(O)R10, -NHC(0)R10, C,-C6 heterocycle having 1 to 4 heteroatoms optionally substituted with 1 to 5 R!", and -C(O)NR,0NHC(O)R10; and
R10 is C1-C12 alkyl;
provided that: for the compound of Formula IA, when R is -OH or C i -C i2 alkyl optionally substituted with 1 to 5 R , then R3 is hydrogen; and
for the compound of Formula IA, when RJ is hydrogen, and -L-R4 is ethyl, then R is not
-OR1.
[0008] This invention is also directed to pharmaceutical compositions comprising a pharmaceutically acceptable excipient and a therapeutically effective amount of a compound of this invention or a mixture of one or more of such compounds.
[0009] This invention is still further directed to methods for treating pain and/or addiction in a patient in need thereof, which methods comprise administering to the patient a one or more of compounds or a pharmaceutical composition of this invention.
[0010] This invention is also directed to methods of inhibiting a3b4 nicotinic acetylcholine receptors.
[0011] This invention is also directed to methods for treating of nicotine addiction and methods for treating of other substance abuse related disorders (SRDs ).
Detailed Description of the Invention
[0012] This invention is directed to ribose substituted noribogaine for use in treating pain and/or addition. However, prior to describing this invention in greater detail, the following terms will first be defined.
[0013] It is to be understood that this invention is not limited to particular embodiments described, as such may. of course, vary. It is also to be understood that the terminology used herein is for the purpose of describing particular embodiments only, and is not intended to be limiting, since the scope of the present invention will be limited only by the appended claims.
[0014J It must be noted that as used herein and in the appended claims, the singular forms "a", "an", and "the" include plural referents unless the context clearly dictates otherwise. Thus, for example, reference to '"a pharmaceutically acceptable excipient" includes a plurality of pharmaceutically acceptable excipients. 1, Definitions
10015] Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. As used herein the following terms have the following meanings.
[0016] As used herein, the term "comprising" or "comprises" is intended to mean that the compositions and methods include the recited elements, but not excluding others.
"Consisting essentially of when used to define compositions and methods, shall mean excluding other elements of any essential significance to the combination for the stated purpose. Thus, a composition consisting essentially of the elements as defined herein would not exclude other materials or steps that do not materially affect the basic and novel characteristic(s) of the claimed invention. "Consisting of shall mean excluding more than trace elements of other ingredients and substantial method steps. Embodiments defined by each of these transition terms are within the scope of this invention.
[0017] The term "about" when used before a numerical designation, e.g., temperature, time, amount, and concentration, including range, indicates approximations which may vary b ( I ) or ( - ) 10 %, 5 % or 1 %.
[0018] Noribogaine can be depicted by the following Formula:
Figure imgf000006_0001
and can be synthesized by the O-demethylation of ibogaine. This may be accomplished, for example, by reacting ibogaine with boron tribromide/methylene chloride at room temperature and then purifying the product using known procedures. Ibogaine may be obtained from natural or commercial sources, or can be synthesized by methods known in the art (see Huffman, et al, J. Org. Chem. 50: 1460 (1 85)). In addition, noribogaine may also be obtained from the National Institute on Drug Abuse (Rockville, d.j.
[001 ] As used herein, the term "hydrolyzable group1' refers to a group that can be hydrolyzed to release the free hydroxy group under hydrolysis conditions. Examples of hydrolvsable group include, but are not limited to those defined for R1 above. Preferred hydrolvsable groups include carboxyl esters, phosphates and phosphate esters. The hydrolysis may be done by chemical reactions conditions such as base hydrolysis or acid hydrolysis or may be done in vivo by biological processes, such as those catalyzed by a phosphate hydrolysis enzyme, Nonlimiting examples of hydrolysable group include groups linked with an cstcr-bascd linker (-C(O)O- or -OC(U)-), an amide-based linker (-C(O)NR40- or -NR40C(O)-), or a phosphate-linker (-P(O)(OR40)-O-, -O-P(S)(OR40)-O-. -0-P(S)(SR40)- 0-, -S-P(0)(OR4f;)-0-. -O-P(O)(OR40)-S-. -S-P(O)(OR40)-S-, -0-P(S)(OR4(')-S-, -S- P(S)(OR40)-O-. -O-P(O)(R, )-O-, -O-P(S)(R 0)-O-, -S-P(O)(R 0)-O-. -S-P(S)(R40)-O-, -S- P(O)(R40)-S-, or -O-P(S (R40)-S-) where R40 can be hydrogen or alkyl.
1002 j "Alkyl" refers to alkyl groups having from 1 to 12 carbon atoms, preferably 1 to 6 carbon atoms and more preferably 1 to 3 carbon atoms. The alkyl group may contain linear or branched carbon chains. This term is exemplified by groups such as methyl, ethyl, n- propyl, iso-propyl, n-butyl, L-butyl. n-pentyl and the like. The term "Cx alkyl" refers to an alkyl group having x carbon atoms, wherein x is an integer, for example. C3 refers to an alkyl group having 3 carbon atoms.
[0021] "Alkenyl" refers to straight or branched hydrocarbyl groups having from 2 to 12 carbon atoms, preferably 2 to 6 carbon atoms and more preferably 2 to 4 carbon atoms and having at least 1 and preferably from 1 to 2 sites of vinyl (>C=C<) unsatiiration. Such groups are exemplified, for example, by vinyl, allyl, and but-3-en- l -yl. Included within this term are the cis and trans isomers or mixtures of these isomers.
[0022] "Alkynyl" refers to straight or branched monovalent hydrocarbyl groups having from 2 to 12 carbon atoms, preferably 2 to 6 carbon atoms and more preferably 2 to 3 carbon atoms and having at least 1 and preferably from 1 to 2 sites of acetylenic (-C≡C-) unsatiiration. Examples of such alkynyl groups include acetylenyl (-C≡CH), and propargyl
(-CH2C≡CH).
[0023] "Alkoxy" refers to the group -O-alkyl wherein alkyl is as defined herein. Alkoxy includes, by way of example, methoxy, ethoxy, n-propoxy, iso-propoxy, n-butoxy, t-butoxy, sec-butoxy, n-pentoxy and the like.
|l)024| "AevT* refers to the groups R4 l -C(0)-, wherein R41 is selected from the group consisting of hydrogen, aikyl, cycloalkyl. aryl, heteroaryl, and heterocyclic, wherein alkyl. cycloalkyl, aryl, heteroaryl, and heterocyclic are as defined herein and are optionally substituted with 1 to 5 substituents independently selected from the group consisting of halo, Ci to C3 alkyl, hydroxy!, and C( to C3 alkoxy. [00251 "Aryl" or "Ar" refers to a monovalent aromatic carbocyclic group of from 6 to 14 carbon atoms having a single ring (e.g., phenyl) or multiple condensed rings (e.g., naphthyl or anthryl) which condensed rings may or may not be aromatic (e.g., 2-benzoxazoIinone, 2H- l ,4-bcnzoxazin-3(4H)-one-7-yl, and the like) provided that the point of attachment is at an aromatic carbon atom.
[0026J "Aryloxy" refers to the group -O-aryl wherein aryl is as defined herein. Aryloxy includes, by way of example, methoxy, ethoxy, n-propoxy. iso-propoxy, n-butoxy. t-butoxy, sec-butoxy, n-pentoxy and the like.
[0027] ""Carboxyl" refers to -COOFI or salts thereof.
[0028] "Carboxyl esters" refers to the groups -C(0)0-R42, wherein R42 is selected from the group consisting of hydrogen, alkyl, cycloalkyl, aryl, heteroaryl, and heterocyclic, wherein alkyl, cycloalkyl, aryl, heteroaryl, and heterocyclic are as defined herein and are optionally substituted w ith 1 to 5 substituents independently selected from the group consisting of halo, C ] to C3 alkyl, hydroxy!, and C| to C alkoxy.
[0029] ""Cycloalkyl" refers to cyclic alkyl groups of from 3 to 10 carbon atoms having single or multiple cyclic rings including, by way of example, adamantyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclooctyl and the like.
[0030] "Halo" or "halogen" refers to fluoro, chloro, bromo and iodo and preferably is fluoro or chloro.
[0031] ""Heteroaryl" refers to an aromatic group of from 1 to 10 carbon atoms and 1 to 4 heteroatoms selected from the group consisting of oxygen, nitrogen, sulfur within the ring, wherein the nitrogen and/or sulfur atom(s) of the heteroaryl are optionally oxidized (e.g., N- oxide, ~S(0)- or -S( ( > >_- >. Such heteroaryl groups can have a single ring (e.g., pyridyl or furyl) or multiple condensed rings (e.g.. indolizinyl or benzothienyl) wherein the condensed rings may or may not be aromatic and/or contain a heteroatom provided that the point of attachment is through an atom of the aromatic heteroaryl group. Examples of heteroaryls include pyridyl, pyrrolyl, indolyl, thiophenyl, and furyl,
[0032] ' Ielerocycle" or "heterocyclic'"' or "heterocycloalky " or "heterocyclyl" refers to a saturated or partially saturated, but not aromatic, group having from 1 to 10 ring carbon atoms and from 1 to 4 ring heteroatoms selected from the group consisting of nitrogen, sulfur, or oxygen. Heterocycle encompasses single ring or multiple condensed rings, including fused bridged and spiro ring systems. In fused ring systems, one or more the rings can be cycloalkyl, ar l, or heteroaryl provided that the point of attachment is through the
non-aromatic heterocyclic ring. In one embodiment, the nitrogen and/or sulfur atom(s) of the heterocyclic group arc optionally oxidized to provide for the N -oxide, sulfinyl, and/or sulfonyl moieties.
[0033] Examples of heteroeycles and heteroaryls include, but are not limited to, azetidine, pyrrole, imidazole, pyrazole, pyridine, pyrazine, pyrimidine, pyridazine, indolizinc, isoindole, indole, dihydroindole, indazole, purine, quinolizine, isoquinoline, quinoline, phthalazine, naphthylpyridine, quinoxaline, quinazoline, cinnoline. pteridine. carbazole, carboline, phenanthridine. acridine. phenanthroline. isothiazole, phenazine, isoxazole, phenoxaziiie. phenothiazine, imidazolidine, imidazoline, piperidine, piperazine, indoline, phthalimide, 1 ,2,3,4-tetrahydro-isoquinoline, 4,5,6,7-tetrahydrobenzo[b]thiophene, thiazole, thiazolidine, thiophene, benzo[b]thiophene. morpholinyl, thiomorpholinyl (also referred to as
thiamorpholinyl). piperidinyl, pyrrolidine, tetrahydrofuranyl, and the like.
[0034] "Alkylene" refers to a divalent radical of an alkyl group. '"Methylene" refers to the group -CH2-.
[0035] "Arylcnc" refers to a divalent radical of an aryl group. "Phenylene" refers to the divalent phenyl group -( '<,! I >,-.
[0036] As used herein, the term "phosphate ester'" refers to any one of the mono-, di- or triphosphate esters of noribogaine, wherein the mono-, di- or triphosphate ester moiety is bonded to the 12-hydroxy group and/or the indole nitrogen of noribogame.
[0037] As used herein, the term "monophosphate" refers to the group -P(0)(OH)2.
[0038] As used herein, the term "diphosphate" refers to the group -P(0)(OH)-OP(0)(OH)2.
[0039] As used herein, the term "triphosphate" refers to the group -P(0)(OH)- (ΟΡ(Ό)(ΟΗ))2ΟΗ.
10040] It is understood that the above definitions are not intended to include impermissible substitution patterns (e.g., methyl substituted with 5 fluoro groups or a hydroxy! group alpha to ethenylic or acetylenic unsaturation). Such impermissible substitution patterns arc well known to the skilled artisan.
[0041 ) The term "pharmaceutically acceptable prodrugs" refers to art recognized
modifications to one or more functional groups which functional groups are metabolized in vivo to provide a compound of this invention or an active metabolite thereof. Such functional groups are well known in the art including acyl groups for hydroxy! and/or amino substitution, esters of mono-, di- and tri-phosphates wherein one or more of the pendent hydroxyl groups have been converted to an alkoxy, a substituted alkoxy, an aryloxy or a substituted aryloxy group, and the like.
[0042] "Pharmaceutically acceptable salt" refers to pharmaceutically acceptable salts, including partial salts, of a compound, which salts are derived from a variety of organic and inorganic counter ions well known in the art and include, by way of example only, sodium, potassium, calcium, magnesium, ammonium, tetraal ky 1 ammoni um , and the like; and when the molecule contains a basic functionality, salts of organic or inorganic acids, such as hydrochloride, hydrobromide. tartrate, mesylate, acetate, lactate, succinate, citrate, salicylate, malale, maleate, oxalate, phosphate, phosphite, sulfate, nitrate, perchlorate, aconitate, thalate, embonate, enanthate, and the like.
[0043] "Pharmaceutically acceptable partial salts" refers to compounds having a substituent capable of having more than one group form a salt but less than the maximum amount of such groups actually form a salt. For example, a diphospho group can form a plurality of salts and, if only partially ionized, the resulting group is sometimes referred to herein as a partial salt.
[0044] As used herein, the term "therapeutically effective amount'' refers to the amount of a composition of this invention that is sufficient to effect treatment, as defined herein, when administered to a subject in need of such treatment. The therapeutically effective amount will vary depending upon the subject and condition being treated, the weight and age of the subject, the severity of the condition, the particular composition or excipient chosen, the dosing regimen to be followed, timing of administration, the manner of administration and the like, all of which can be determined readily by one of ordinary skill in the art.
[0045] As used herein, the term "treatment" or "treating" means any treatment of a disease or condition in a patient, including:
• preventing or protecting against the disease or condition, that is, causing the clinical symptoms not to develop, for example, in a subject at risk of suffering from such a disease or condition, thereby substantially averting onset of the disease or condition;
• inhibiting the disease or condition, that is, arresting or suppressing the development of clinical symptoms; and/or • relieving the disease or condition that is, causing the regression of clinical symptoms,
[0046] As used herein, the term "pain" refers to all types of pain, including neuropathic and nociceptive pain. It is also contemplated that the compositions disclosed herein can be used to treat other types of pain such as phantom pain which is the sensation of pain from a limb or organ that has been lost or from which a person no longer receives physical signals, and is an experience almost universally reported by amputees and quadriplegics.
(0047J As used herein, the term "'addiction" refers to a persistent behavioral pattern marked by physical and/or psychological dependency to a substance, particularly drugs such as narcotics, stimulants, and sedatives, including but not limited to heroin, ***e, alcohol, nicotine, caffeine, amphetamine, desoxyephedrine, methadone and combinations thereof. As used herein, the "treatment of addiction in a patient" refers to reducing the withdrawal symptoms associated with drug dependency as well as alleviating drug cravings in addicts. Such symptoms include nausea, vomiting, anxiety, abdominal cramps, muscle pain, chills and headache.
100481 As used herein, the term "patient" refers to mammals and includes humans and non-human mammals.
2. Compounds
[0049] This invention is relates to noribogaine derivative compounds. Such compounds are contemplated as being useful in treating pain and/or drug dependency. Accordingly, in one of its composition aspects, this invention is directed to a compound or a pharmaceutically acceptable salt thereof wherein said compound is represented by Formula IA or Formula IB, or a pharmaceutically acceptable salt of each thereof:
Figure imgf000011_0001
IB
W hc- 6i i
R is OR1 or optionally substituted with 1 to 5 R9;
R1 is selected from the group consisting of hydrogen, -C(0)OX, -SO2OR10, a
monophosphate, a diphosphate, a triphosphate, and ~C(Q)N(Y)2 where X is C i -C alkyl optionally substituted with 1 to 5 R9, and each Y is independently selected from the group consisting of hydrogen, CVC6 alkyl optionally substituted with 1 to 5 R9, C-6-Ci aryl optionally substituted w7ith 1 to 5 RJ, C3-C 1 cycloalkyl optionally substituted with 1 to 5 R9. Ci - | 0 heteroaryl having 1 to 4 heteroatoms and which is optionally substituted with 1 to 5 R9, Ci-C 10 heterocyclic having 1 to 4 heleroatoms and which is optionally substituted with 1 to 5 R9, or where each Y, together with the nitrogen atom bound thereto form either a CrQ heterocyclic having 1 to 4
heteroatoms and which is optionally substituted with 1 to 5 R9, or a C| -Cf, heteiOaryl having 1 to 4 heteroatoms and which is optionally substituted with 1 to 5 R9;
R' is halo, OR20, or Cj-C alkyl optionally substituted with 1 to 5 R9:
R20 is selected from the group consisting of hydrogen , -C(0)X, -C(0)OX and -C(0)N(Y)2 where X and Y are defined as above;
R2 is hydrogen, -SO2OR10, a monophosphate, a diphosphate, or a triphosphate;
R3 is selected from the group consisting of hydrogen, -(CH2)mOR7, -CR6(OH)R7. -(CH2)mCN, -(CH2)mCOR7, -(CH2)mC02R7, -(CH2)mC(0)NR6R7, -(( 1 l; ),„C'( ( ) )N ' N R R .
-(CH2)mC(0)NR6NR7C(0)R8 ; and -(CH2)mNR6R7;
m is 0, 1 , or 2;
L is a bond or C i -C 12 alkylene;
R4 is selected from the group consisting of C 1 -C 12 alkyl substituted with 1 to 5 R9, C2-Ci2 alkenyl substituted with 1 to 5 R9, -X2-R6, -(X2-Y2)n-X2-R6, -S02NR6R7, -0-C(0)R8, -C(0)OR7, -C(0)NR6R7, -NR6R7 ; -NHC(0)R8, and -NR6C(0)R8;
X2 is selected from the group consisting of O and S;
Y2 is C 1 -C4 alkylene or C C 10 arylene, or a combination thereof;
n is i , 2. or 3:
Rb and R' are each independently selected from the group consisting of hydrogen. C|-Ci2 alkyl optionally substituted with 1 to 5 R9, ( Cs heterocyclic having 1 to 4
heteroatoms which is optionally substituted with 1 to 5 R9. C3-C10 cycloalkyl optionally substituted with 1 to 5 R9. Q-Cio aryi optionally substituted with 1 to 5 R9 and C : - {, heteroaryl having 1 to 4 heteroatoms optionally substituted with 1 to 5 R9; or R6 and R' are joined to form CyCeheterocyele having 1 to 4 heteroatoms which is optionally substituted with 1 to 5 R9:
R8 is selected from the group consisting of C]-C 12 alkyl optionally substituted with 1 to 5 R9, Cs-Cehelerocycle having 1 to 4 heteroatoms optionally substituted with 1 to 5 R9, Cj- Ciocycloalkyl optionally substituted with 1 to 5 R9, C6-Ci aryl optionally substituted with 1 to 5 R9 and C]-C6heteroaryl having 1 to 4 heteroatoms optionally substituted with 1 to 5 R9;
R" is selected from the group consisting of C1-C4 alkyl, phenyl, halo, -OH, -OR10, -CN, -COR10, -CO2R10 ; -C(0)NHR10, -NR10R10, -C(O)NR,0R10. -C(0)NHNHR10.
-C(O)NRI0NHR10, -QO^R'WR10, -C(0)NHNRIOC(0)R,(),
-C(0)NHNHC(0 R'"; -SO2NRL0R10, -C(0)NR10NRIOC(0)R10, -NHC(0)R10, CI-C6 heterocycle having 1 to 4 heteroatoms optionally substituted with 1 to 5 R10, and -C(O)NR10NHC(O)R10; and
R10 is C1-C12 alkyl;
provided that:
for the compound of Formula IA, when R is -OH or C1-C12 alkyl optionally substituted with 1 to 5 R9, then R"5 is hydrogen; and
for the compound of Formula IA, when R"1 is hydrogen, and -L-R4 is ethyl, then R is not
-OR1.
[0050] In one embodiment, the compound of Formula IA or IB is represented by Formula IA' and IB" respectively:
Figure imgf000013_0001
1 A' IB*
wherein R, R RJ and R4 are as described for formula I.
[0051] In one embodiment, R is halo, such as fluoro or chloro.
[0052] In one embodiment. R is CrC12 alkyl optionally substituted with 1 to 5 R9. In some embodiments, R is C1-C12 alkyl optionally substituted with a -S02NR6R' group. [0053] In another embodiment, R is OR1 , In some embodiments, R1 is selected from the group consisting of C(0)Rs; -C(0)NR5R:> and -C(0)QR5; where each R5 is independently selected from the group consisting of hydrogen, alkyl, alkenyl. alkynyl, aryl. heteroaryl, and hctcrocycle, and wherein the alkyl. alkenyl. alkynyl, aryl. heteroaryl, and heterocycle are optionally substituted with 1 to 5 R .
[0054] In another embodiment, R1 is -C(0)R\ and wherein R5 is Cj-Ci? alkyl or substituted C -C 12 alkyl or aryl. which C i to C alkyl or aryl is optionally substituted with C | to C ¾ alkyl, Ci to C3 alkoxy or aryl. In some embodiments, R is Cj-Q alkyl.
[0055] In some embodiments, R5 is selected from the group consisting of methyl, ethyl, n-propyl, n-butyl, n-pentyl, n-hexyl, n-heptyl. n-octyl, n-nonyl, n-decyl. n-undecyl, n- dodecyl, iso-propyl sec -butyl, tcrt-butyl, phenyl, benzyl, methylbcnzyl, and methoxybenzyl.
[0056] In some embodiments, R1 is benzoyl.
[0057] In some embodiments. R1 is hydrogen.
[0058] In some embodiments, R2 is hydrogen. In some embodiments, R2 is (>(.* : alkyl optionally substituted with 1 to 5 R9, for example, CHiOH. In some embodiments, R2 is aryl optionally substituted with 1 to 5 R9, for example, phenyl substituted with a halo group. In some embodiments, R2 is -C(0)R\ such as -C(0)CH2CH2N(CI l3)2. In some embodiments, R2 is -C(0)NR5R5, such as -C(0)NHCH3 or C(0)NHCH2CH:N(CH3)2. In some
embodiments, R2 is -C(0)OR5. for example, -C(0)OCH3 or -C(0)OCH2CH2N(CH3)2.
[0059] In some embodiments. R is hydrogen or -CHbOH. In some embodiments, R is
[0060] In some embodiments, L-R4 is not ethyl. In some embodiments, L-R 1 is not alkyl. In some embodiments, when R is OR1, L-R4 is not ethyl. In some embodiments, when R is OR1 , L-R'1 is not alkyl.
[0061 ] In some embodiments, L is Cs-CV,aIkyIene,
[0062 J In some embodiments, R4 is -X-R ',
[0063] In some embodiments, X is O,
[0064| In some embodiments. R4 is selected from the group consisting of hydrogen, Ci -Cizalkyl, -CH2Ph, and -C(0)OR7.
[0065] In some embodiments, R4 is -NH2, -CILOCFf CHiOCLL or -OCH3. J 066J In some embodiments, the compound is selected from the group consisting of:
Figure imgf000015_0001
Figure imgf000016_0001

Figure imgf000017_0001
Figure imgf000018_0001
or a pharmac eutical ly acceptable salt thereof.
100671 In a further embodiment, this invention is directed to a compound or a pharmaceutically acceptable salt thereof wherein said compound is represented by Formula 1IA or Formula IIB, or a pharmaceutically acceptable salt of each thereof:
Figure imgf000018_0002
11 A IIB wherein
R11 is halo, -OH, -SH, -NH2, -S(0)2N(R16)2, -X'-L'-R17, -X'-L'-R18, -X'-lJ-R19, or - X'-L'-CHR17R18, where X1 is (>. S or NR16, or -O-Q, where Q is -S02-(C,-Ci: alkyl). a monophosphate, a diphosphate, or a triphosphate;
L1 is alkylene, arylene, -C(0)-alkylene, -C(0)-arylene, -C(0)0-arylene, -C(0)0- alkylene, -C(0)NR19-alkylene, -C(0)NR19-arylene, -C(NRl9)NR,9-alkylene or
-C(NRL9)NR19-arylene, wherein L1 is configured such that -O-L'-R' ' is - OC(0)-alkylene-R17, -OC{0)0-arylene-RN, -OC(0)0-alkylene-R17, -OC(O)- arylene-R17, -OC(0)NRI9-alkylene-R17, -OC(0)NRl9-arylene-R17, - OC( RL9)NRl9-alkylene-R17 or -OC(NR19)NRl9-arylene-R17, and wherein the alkylene and arylene are optionally substituted with 1 to 2 R15;
R12 is hydrogen, -S(()): ( ) ". -S(0)2R19, -C(0)R14, -C(0)NR14R14, -C(0)OR14, CRC12 alkyl optionally substituted with 1 to 5 R15, Ci-Ci2alkenyl optionally substituted with 1 to 5 R15, aryl optionally substituted with 1 to 5 R , -S02- (Ci-Ci2 alkyl), a monophosphate, a diphosphate, or a triphosphate;
R13 is hydrogen, halo, -OR16, -CN, (. Ί -( Ί 2 alkyl, C]-Ci2 alkoxy, aryl or aryloxy, where the alkyl, alkoxy, aryl, and aryloxy are optionally substituted with 1 to 5 R15; each R14 is independently selected from the group consisting of hydrogen, C1-C12 alkyl, C2-Ci2alkenyl, C2-Ci2alkynyl, aryl, heteroaryl, and heterocycle, and wherein the alkyl, alkenyl, alkynyl, aryl, heteroaryl, and heterocycle are optionally substituted with 1 to 5 R13;
R1" is selected from the group consisting of phenyl, halo, -OR16, -CN, -COR15,
-C02R16, -NRI6R16, - NRL6C(0)R16, - NRL6S02R16, -C(0)NRL6R16, -C(0)NRL6NR16R16, -S02NRL6R16 and -C(0)NR16NRL6C(0)R16; each R16 is independently hydrogen or C1-C12 alkyl optionally substituted with from 1 to 3 halo;
R17 is hydrogen, -C(0)R19, ^C(0)OR19, -C(0)N(R,9)2 or -N(R,9)C(0)R!9;
R18 is hydrogen, -N(RI9)2, -C(0)N(RL9)2, -C(NR19)N(RI9)2, -C( S02R19)N(Rl9)2, -NRl9C(0)N(R19)2, -NR,9C(S)N(R,9)2, -NRI9C(NR,9)N(RI9)2,
-NR19C(NS02RI9)N(R19)2 or tetrazole; and each R19 is independently selected from the group consisting of hydrogen, C]-C|2 alkyl and aryl;
provided that:
for the compound of Formula IIA :
when Ri 2 and R13 are hydrogen, then R1 1 is not hydroxy:
when R13 is hydrogen, R is -0-L!-R, 7 5 -O-L'-R1 8, -O-L'-R19, and L1 is
alkylene, then -O-L' -R1 7. -O-L'-R18, -O-L'-R19 are not methoxy; and when R13 is hydrogen, X1 is O, L is -C(0)-alkylene, -C(0)-arylene, -C(0)0- arylene. -C(0)0-alkylene, -C(0)NR19-alkylene, or -C(0)NR, 9-arylene, then none of R1 7, R18 or R19 are hydrogen.
[0068] In one embodiment, the compound of Formula II is represented by Formula IIC or Formula IID, or a pharmaceutically acceptable salt of each thereof:
Figure imgf000020_0001
[0069] In one embodiment of the compounds of Formula IIA, IIB, IIC, or IID, R is halo. In certain embodiments of the compounds of Formula IIA, IIB, IIC, or IID. R 1 1 is bromo. In certain embodiments, R1 1 is chloro. In certain embodiments of the compounds of Formula IIA, UB, IIC, or IID, R 1 1 is fluoro. In certain embodiments, R1 1 is iodo.
[0070] In one embodiment of the compounds of Formula IIA, IIB. IIC, or IID, R1 ' is -OH, - O-L ' -R1 7, -O-L' -R1 8, -O-L' -R19 or -0-L'-CHR1 7R, s. In one embodiment of the compounds of Formula I or IIA I, R1 1 is -SFI, -S-L'-R1 ', -S-L'-R18, -S-L'-R19 or -S-L'-CHR^R18. In one embodiment of the compounds of Formula 11 or IIA, Ru is -NI 12, -NR 16-L!-R17, -NRI 6-L'- R 18, -NR^-L'-R19 or -NR16-L' -CHRI 7R18. In one embodiment of the compounds of Formula IIA, IIB, IIC, or I!D. Rn is ^S(0)2N(R16)2.
|0071] In one embodiment of the compounds of Formula IIA, IIB. IIC, or 11D, 1 1 is -CI, - Br, -I, -OC(0)CH2CHj, -OC(0)CH2Ph, -OC(0)OCti2Cll3, -OC(0)OCI FPh,
-OC(0)NH(CM(CM3)Ph), -OC(NPh)NHCH2CH3, -OC(0)NHCH2Ph, -NH2, -
NHC(0)C(CH3)3, -NHC(0)CF3, ~NHC(0)CH2Ph, -NHC(0)OCH2Ph, -NHC(0)Nl I(Cl I(CII3)Ph); -SI I , -SC(0)OCH2Ph, -SC(0)NH(CH(CH3)Ph), - SC(O)CH2CH20P(0)(OH)2, -OC(0)CH2CH2CH(NH2)(CO2H), -0-(2-OH- C6H3)CH2CH(NH2)(C02H) or -NHC(0)CH2CH2CH(NH2)(C02H).
10072] In one embodiment of the compounds of Formula 1IA, IIB, IIC. or IID, R12 is hydrogen.
1 073] In one embodiment of the compounds of Formula HA, IIB, IIC, or IID, R12 is - CH2CFI3, -CH2CHCH2, -CH2Ph, -C(0)0(CH2)2N(CH3)2 or -C(0)CH2(CH2)2S02N(CH3)2.
[0074] In one embodiment of the compounds of Formula IlA. IIB, IIC, or IID, R' is hydrogen.
[0075] In one embodiment of the compounds of Formula I IA, IIB, IIC, or IID, R3 is halo. In certain embodiments of the compounds of Formula ILA, IIB, IIC, or IID, R3 is bromo. In certain embodiments of the compounds of Formula IIA, IIB. IIC, or IID, R3 is iluoro.
[0076| In one embodiment of the compounds of Formula IIA, IIB, IIC. or IID, R' is -OCH3. -OCH2CH3. -OCFhPh or -CN.
[0077] In some embodiments of the compounds of Formula IIA, IIB, IIC, or IID, the compound is represented by formula HE selected from Table 1 :
Figure imgf000021_0001
HE
Table 1
Figure imgf000021_0002
Figure imgf000022_0001
Figure imgf000023_0001
or a p armaceutca y accepta e sat t ereo .
[0078] In some embodiments of the compounds of Formula IA, the compound is represented by Formula IC selected from Table 2:
Figure imgf000023_0002
able 2
Figure imgf000023_0003
Figure imgf000024_0001
[0079] In certain embodiments, the compound of Formula IA, IB, IC, IIA, IIB, IIC, or IID, is the hydrochloride salt. In another embodiment, the compound of Formula IA, IB, IC, IIA, IIB, IIC, or IID, is the hydrobromide salt. In another embodiment, the compound of Formula IA, IB, IC, IIA, IIB, IIC, or IID, is the phosphate salt. In another embodiment, the compound of Formula IA, IB, IC, IIA, IIB, IIC, or IID, is the sulfate salt.
3. Methods of Use
Treatment of Pain
[0080] In one of its method aspect, the present invention is directed to a method for treating a pain in a patient which method comprises administering to said patient a compound of this invention or a pharmaceutically acceptable salt thereof or a pharmaceutical composition comprising a compound of this invention and a pharmaceutically acceptable excipient. The pain can be any type of pain including, but not limited to neuropathic or nociceptive pain, and various types thereof including somatic, visceral and phantom pain.
Treatment of Addiction
[0081] In another of its method aspect, the present invention is directed to a method for treating addiction in a patient which method comprises administering to said patient a compound of this invention or a pharmaceutically acceptable salt thereof or a composition comprising a compound of this invention and a pharmaceutically acceptable excipient.
[0082] In certain embodiments, the treatment of addiction in a patient comprises alleviating the symptoms associated with withdrawal from drug dependency. Such symptoms include nausea, vomiting, anxiety, abdominal cramps, muscle pain, chills and headache. In addition, it is contemplated that treatment with a compound of this invention decreases the drug cravings normally experienced by addicts after cessation of the self administration of the abused substance. It is contemplated that the compositions disclosed herein are especially useful in the treatment of addiction to narcotics such as heroin and methadone. However, it is also useful in treating patients addicted to ***e, alcohol, amphetamines and combinations of these drugs.
[0083] The invention is also directed to a method for treating drug addiction (involving drug dependency or drug abuse) during withdrawal therapy by administering a compound of this invention to a patient at a dosage sufficient to reduce or eliminate one or more symptoms associated with withdrawal. Such symptoms include nausea, vomiting, anxiety, abdominal cramps, muscle pain, chills and headache. In addition, treatment with a compound of this invention is contemplated to decrease the drug cravings normally experienced by addicts after cessation of the self administration of the abused substance, for example, narcotics such as heroin and methadone. However, compounds of this invention are contemplated to be also useful in treating patients addicted to ***e, alcohol, amphetamines and combinations of these drugs. Compounds of this invention may be administered to patients suffering from drug dependence or abuse in conjunction with an opioid antagonist such as naloxone, naltrexone or nalorphine, for example, at a concentration of between 0.1 5 mg and 0.5 mg for each mg of the compound of this invention administered.
Treatnmeni of nicotine addiction
[0084] Tobacco use remains a major health problem despite widespread knowledge of the damaging consequences. Although current smoking cessation therapies, including nicotine replacement, bupropion, and varenicline, have had some success, they are inadequate since only a fraction (30-40%) of smokers who try these medications abstain from tobacco use. Consequently, more effective or add-on therapies are needed. Nicotine is one of the most addictive drugs that is widely used; 95% or more of its users with a strong desire to stop using relapse within one year. Chronic nicotine use leads to physiological changes in nAChR function and expression, including upregulation of high-affinity a4b2 nAChRs expression and reduced receptor function due to desensitization. in addition to the reinforcing properties of nicotine, the physical discomfort associated with nicotine withdrawal includes severe cravings, anxiety, dysphoria, and autonomic dysfunction. Although it is known that the reinforcing and addictive eff ects of nicotine are due to is actions on nAChRs, the
identification of which nAChR subtypes play a role in the various aspects of nicotine dependence/withdrawal, and their validation as drug targets for medication development remains a complex area of investigation. The a4 subunit is implicated in nicotine reinforcement, sensitization and the development of tolerance. While the development of tolerance is not regulated by a7 nAChRs, a recent stud indicated that that these receptors may control the severity of the withdrawal symptoms. Thus, there is a complex regulatory interplay of nAChRs, which likely contribute to nicotine dependence/withdrawal.
[0085] Neuronal nicotinic acetylcholine receptors (nAChRs) have been a target for CNS drug discovery efforts for the past two decades. Mounting evidence suggests that the addictive effects of nicotine, occur through interaction with its receptor in the mesolimbic dopamine system. However, the molecular identity of the nicotinic receptors responsible for drug seeking behavior, their cellular and subcellular location and the mechanisms by which these receptors initiate and maintain nicotine addiction are poorly defined. While nicotine and related natural products have been used for smoking cessation in various formulations (e.g., gum, spray, patches), it was only in 2006 with Pfizer' s launch of varenicline
(ChantixTM) for smoking cessation that a new chemical entity ( NCI ) targeting neuronal AChRs was approved for this condition. Varenicline is a weak partial agonist for a4b2 nAChR subtypes and a less potent agonist at a7 subtypes. A serendipitous finding in aiding smoking cessation is the discovery that the atypical anti-depressant bupropion, whose mechanism of action as a non-competitive antagonist of a3b4 nAChRs and antidepressant activity underlie its efficacy. Buproprion analogs are under development as potential pharmacotherapies for smoking cessation. Second generation noribogaine analogs targeted to a3b4 nAChRs offer a novel synthetic route for development of potential small molecules to treat tobacco addiction and other SRDs.
[0086] Recent genetic association studies show that single nucleotide polymorphisms
(SNPs) in the gene cluster CHRNA5/A3/B4, encoding for the a3, a5 and b4 nAChR subunits are associated with increased risk for heavy smoking, inability to quit, and increased sensitivity to nicotine. Furthermore, a3b4 nAChRs or other b4 containing receptors have been suggested to be involved in nicotine withdrawal. The expression of the a3b4 AChR is restricted to a few discrete brain areas, including the medial habenula and interpeduncular nucleus, and autonomic ganglia. Recent behavioral studies in genetic mouse models suggest that disrupted function and/or expression of the a3b4 (and a5 subunits) in the habenulo-IPN tract may be critical for regulating nicotine intake and the addictive properties of this substance. The habcnula-intcrpeduncular system and a3b4 nAChR subunits in this pathway play an important role in nicotine withdrawal. The compounds of this invention were found to inhibit a3b4 nAChRs.
[0087) In another of its method aspect, the present invention is directed to a method of treating nicotine addiction in a patient which method comprises administering an effective amount of a compound of this invention or a pharmaceutically acceptable salt thereof or a composition comprising a compound of this invention and a pharmaceutically acceptable excipient to a patient in need thereof.
[0088] In another of its method aspect, the present invention is directed to a method of inhibiting a3b4 nicotinic acetylcholine receptors which method comprises administering an effective amount of a compound of this invention or a pharmaceutically acceptable salt thereof or a composition comprising a compound of this invention and a pharmaceutically acceptable excipient.
Combination therapy
[0089] Compounds of this invention maybe used alone or in combination with other compounds to treat pain and/or addiction. When administered with another agent, the coadministration can be in any manner in which the pharmacological effects of both are manifest in the patient at the same time. Thus, co-administration does not require that a single pharmaceutical composition, the same dosage form, or even the same route of administration be used for administration of both the compound of this invention and the other agent or that the two agents be administered at precisely the same time. However, coadministration will be accomplished most conveniently by the same dosage form and the same route of administration, at substantially the same time. Obviously, such administration most advantageously proceeds by delivering both active ingredients simultaneously in a novel pharmaceutical composition in accordance with the present invention.
[0090] In some embodiments, a compound of this invention can be used as an adjunct to conventional drug withdrawal therapy, specifically providing for the administration of a compound of this invention with one or more opioid antagonists.
4, Compositions
[00911 In another aspect, this invention is also directed to pharmaceutical compositions comprising a pharmaceutically acceptable excipient and a compound of this invention or mixtures of one or more of such compounds. [0092 ] Although compositions suitable for oral, intravenous or intraarterial delivery will probably be used most frequently, other routes that may be used include peroral, pulmonary, rectal, nasal, vaginal, lingual, intramuscular, intraperitoneal, intracutaneous and subcutaneous routes. In addition, it is contemplated that the composition can be administered transdermally in which drug is applied as part of a cream, gel, or patch (for examples of transdermal formulations, see U.S. Pat. Nos. 4,806,341 ; 5, 149,538; and 4,626,539). Other dosage forms include tablets, capsules, pills, powders, aerosols, suppositories, parenterals, and oral liquids, including suspensions, solutions and emulsions. Sustained release dosage forms may also be used, for example, in a transdermal patch form. All dosage forms may be prepared using methods that are standard in the art (see e.g., Remington's Pharmaceutical Sciences, 16th ed., A. Oslo editor, Easton Pa. 1980). Intranasal administration is an effective method for delivering a therapeutic agent directly to the respiratory tract, where the therapeutic agent may be quickly absorbed.
[0093] The compositions are comprised of in general, a compound of this invention or a mixture thereof in combination with at least one pharmaceutically acceptable excipicnt. Acceptable excipients are non-toxic, aid administration, and do not adversely affect the therapeutic benefit of the compound of this invention. Such excipients may be any solid, liquid, semi-solid or, in the case of an aerosol composition, gaseous excipient that is generally available to one of skill in the arl. Pharmaceutical compositions in accordance with the invention are prepared by conventional means using methods known in the art.
[0094J Solid pharmaceutical excipients include starch, cellulose, talc, glucose, lactose, sucrose, gelatin, malt, rice, flour, chalk, silica gel, magnesium stearate, sodium stearate, glycerol monoslearate, sodium chloride, dried skim milk and the like. Liquid and semisolid excipients may be selected from glycerol, propylene glycol, water, ethanol and various oils, including those of petroleum, animal, vegetable or synthetic origin, e.g., peanut oil, soybean oil, mineral oil, sesame oil, etc. Preferred liquid carriers, particularly for injectable solutions, include water, saline, aqueous dextrose, and glycols.
[0095| Compressed gases may be used to disperse a compound of this invention in aerosol form. Inert gases suitable for this purpose are nitrogen, carbon dioxide, etc. Other suitable pharmaceutical excipients and their formulations are described in Remington's
Pharmaceutical Sciences, edited by E. W. Martin (Mack Publishing Company, 18th ed.,
1990), [0096] The compositions disclosed herein may be used in conjunction with any of the vehicles and cxcipicnts commonly employed in pharmaceutical preparations, e.g., tale, gum arable, lactose, starch, magnesium stearate, cocoa butter, aqueous or non-aqueous solvents, oils, paraffin derivatives, glycols, etc. Coloring and flavoring agents may also be added to preparations, particularly to those for oral administration. Solutions can be prepared using water or physiologically compatible organic solvents such as ethanol, 1 ,2-propylene glycol, polyglycols, dimethylsulfoxide. fatty alcohols, triglycerides, partial esters of glycerine and the like. Parenteral compositions containing noribogaine may be prepared using conventional techniques that may include sterile isotonic saline, water, 1 ,3-butanediol, ethanol, 1 ,2- propylene glycol, polyglycols mixed with water, Ringer's solution, etc.
[0097] The amount of the compound in a formulation can vary within the full range employed by those skilled in the art. Typically, the formulation will contain, on a weight percent (wt %) basis, from about 0.01 to 99.99 wt % of a compound of this invention based on the total formulation, with the balance being one or more suitable pharmaceutical excipients. Preferably, the compound is present at a level of about 1 to 80 wt %. In a liquid composition, a compound of this invention should generally be present in such compositions at a concentration of between about 0.1 and 20 mg/ml. When either naloxone or naltrexone is combined with a compound of this invention, they should be present at 0.05 to 0.5 mg for each mg of the compound of this invention.
[0098] The choice of formulation depends on various factors such as the mode of drug administration and bioavailability of the drug substance. For delivery via inhalation the compound can be formulated as liquid solution, suspensions, aerosol propellants or dry powder and loaded into a suitable dispenser for administration. There are several types of pharmaceutical inhalation devices-nebulizer inhalers, metered dose inhalers (MDI) and dry powder inhalers (DPI). Nebulizer devices produce a stream of high velocity air that causes the therapeutic agents (which are formulated in a liquid form) to spray as a mist that is carried into the patient's respiratory tract. MDI's typically are formulation packaged with a compressed gas. Upon actuation, the device discharges a measured amount of therapeutic agent by compressed gas. thus affording a reliable method of administering a set amount of agent. DPI dispenses therapeutic agents in the form of a free flowing powder that can be dispersed in the patient's inspiratory air-stream during breathing by the device. In order to achieve a free flowing powder, the therapeutic agent is formulated with an excipient such as lactose. A measured amount of the therapeutic agent is stored in a capsule form and is dispensed with each actuation.
[0099] Recently, pharmaceutical formulations have been developed especially for drugs that show poor bioavailability based upon the principle that bioavailability can be increased by increasing the surface area i.e., decreasing particle size. For example, U.S. Patent No. 4,107,288 describes a pharmaceutical formulation having particles in the size range from 10 to 1 ,000 mn in which the active material is supported on a crosslinked matrix of
macromolecules. U.S. Patent No. 5,145,684 describes the production of a pharmaceutical formulation in which the drug substance is pulverized to nanopartieles (average particle size of 400 nm) in the presence of a surface modifier and then dispersed in a liquid medium to give a pharmaceutical formulation that exhibits remarkably high bioavailability.
[0100] Generally, the compound of this invention is administered in an effective amount. It is contemplated that the dosage required for treating pai n or addition, or a combination thereof may differ according to the condition(s) being treated, however, the dosing regimen can be readily determined by the attending clinician based on the desired treatment. It is contemplated that generally, the dosage of a compound of this invention administered to a patient may be from about 0.01 to about 1000 mg per kg of body weight per day (mg/kg/day), or from 0.05 to 500 mg/kg/day, preferably, from about 0.1 to about 100 mg/kg/day, more preferably from about 0.5 to 50 mg/kg/day. For example, for administration to a 70 kg person, the dosage range would preferably be about 35 to 700 mg per day.
[0101] In addition to the methods discussed above, the present invention is directed to a pharmaceutical composition, preferably in unit dose form, comprising a compound of this invention. When administered to a patient, one or more unit doses provide an amount of a compound of this invention effective to treat pain and/or addition.
[0102] The amount of the composition administered will depend on a number of factors, including but not limited to the desired final concentration of the compound, the
pharmacokinetic and pharmacodynamic properties of the compound, the size, age, and physiological profile of the patient, and the like. The active compound is effective over a wide dosage range and is generally administered in a pharmaceutically effective amount. It will be understood, however, that the amount of the compound actually administered will be determined by a physician, in the light of the relevant circumstances, including the condition to be treated, the chosen route of administration, the actual compound administered, the age, weight, and response of the indi vidual patient, the severity of the patient's symptoms, and the like.
[0103] Determination of dosages is well within the empiric knowledge of persons skilled in the art; nonetheless, it can be appreciated that estimates of final dosages can be made by approximating the concentration of compound necessary to achieve a desired therapeutic activity, such as treatment of pain and/or addiction. Extrapolation to a specified mammal ian dosage range, or more particularly a human dosage range is well w ithin the skill of the practitioner.
[0104] In some embodiments, compositions are administered in one dosing of a single formulation and in other embodiments, compositions are administered in multiple dosing of a single formulation within a specified time period. In some embodiments, the time period is between about 3 hours to about 6 hours. In other embodiments, the time period is between about 6 hours and 12 hours. In additional embodiments, the time period is between about 12 hours and 24 hours. In yet further embodiments, the time period is between about 24 hours and 48 hours. The administration of separate formulations can be simultaneous or staged throughout a specified time period, such that all ingredients are administered within the specified time period.
[0105] The following are examples of the compositions of this invention.
Example 1 : Tablet formulation
|0106] 'The following ingredients are mixed intimately and pressed into single scored tablets.
Figure imgf000031_0001
Example 2; Capsule formulation
[0107] The following ingredients are mixed intimately and loaded into a hard-shell gelatin capsule.
Figure imgf000032_0001
Example 4: Suspension formulation
[0108] The following ingredients are mixed to form a suspension for oral administration (q.s. = sufficient amount).
Figure imgf000032_0002
Example 3: Injectable formulation
[0109] The following ingredients are mixed to form an injectable formulation.
Ingredient Quantity per tablet, mg Compound of Formula I 0.2 mg-20 mg sodium acetate buffer solution, 0.4 M .0 mL
IK I (IN) or NaOH (IN) q.s. to suitable pH water (distilled, sterile) q.s. to 20 mL
Example 5: Suppositor formulation
[0110] A suppository of total weight 2.5 g is prepared by mixing the compound invention with Witepsol® H- 15 (triglycerides of saturated vegetable fatty acid;
Riches-Nelson, Inc., New York), and has the following composition:
Figure imgf000033_0001
Example 6: Screening compounds for nAChR target
[0111] Preliminary functional studies performed by Caliper Life Sciences (Hopkinton, MA), using the FLIPR® Ca+2 flux assay, demonstrated that noribogaine (12- hydroxyibogamine) was more potent as an antagonist (IC50 - 462 nM) for epibatine induced CaT2 flux in SH-SY5Y cells compared to 18-MC (3.89 μΜ), where SH-SY5Y cells predominantly express the α3β4 nicotinic receptor subtype. Additional functional studies using HE 293 cells expressing α3β4 nicotinic receptors show that congeners of 18-MC, with identical substituents as shown above, can modulate the rate and recovery of nicotinic receptor desensitization. The FIJPR® Ca+2 flux assay is used with IIEK293 cells over- expressing a3b4, a4b2 or a7 to determine the potency and selectivity of the noribogaine congeners. The HEK293 (a3b4, a4b2 and a7) and SH-SY5Y cell lines (a3b4) expressing nAChRs are used. Validated assays optimized with respect to sensitivity, dynamic range, signal intensity and stability for chemical probe validation and SAR refinement are used.
[0112] Nicotinic functional assays are performed using the Molecular Devices FLIPR® Calcium 5 Assay Kit. HEK293 cells overexpressing α3β4, σ.4β2 or u7 (50,000 cells per well in 100 μΐ growth media) and are plated in clear bottom black walled 96-well microplates and incubated for 24-48 hours at 37°C, 5% C02 until a 80-90% confluent monolayer is formed. On the day of the assay, 100 μΐ of Calcium 5 Dye is added to each well. Following addition of dye. cells are incubated at 25°C for 60 minutes. Following dye loading, 50 μΐ of a 5x solution of reference or test compound of appropriate concentration range (0.01 - 30 μΜ), prepared in 20 mM HEPES pH 7.4 in HBSS. is added to each well. Plates are then transferred to the Molecular Devices Flexstation® which transfers 50 of a 5x solution (500 n ) of epibatine to each well to initiate the Ca+2 flux. Additional wells (in quadruplicate) are prepared with no added test or reference ligand for measurement of 100% flux. The RFU (relative fluorescence units) in the presence of the test or reference compounds is then expressed on the percent scale. The percent inhibition of epibatidine induced Ca+2 flux is calculated as follows: % inhibition = 100% - %RFU (relative fluorescence units). The IC50 values for reference and test compounds are obtained by fitting normalized RFU data using Graphpad Prism software.
[0113] 86RB+ efflux assays are also used to assess whether noribogaine or its analogs have activity as antagonists at human nAChR using naturally or heterologously expressed cell lines and compared to synaptosomal preparations from mouse brain. SHSY cells function only for measures of a3b4, because a7 nACliRs, which are also expressed by these cells inactivate too quickly to detectably contribute to ion influx. 86RB+ efflux in the presence of the compounds is used to test for agonist and antagonist activity at hAChRs. Representative concentration curves are obtained to complete the SAR profiling of the compound series. Inhibition of agonist stimulated DA and ACh release are tested in striatal and IPN synaptosomal preparations according to known methods using L-nicotine or cy tosine to stimulate release, respectively. IC50 values for inhibition of ACh and DA release (μΜ) are determined from the curve fits of the data with or without test compound present.
Table 3: Inhibition of epibatidine-induced calcium response in a3b4 nAChRs-containing cell lines
Figure imgf000034_0001
[0114] The results shown in Tables 3 and 4 reveal that addition of a substituent R 1 at C- 1 8 C(0)0(CH2)2 leads to higher affinity for the receptor relative to the unsubstituted parent compound. The highest effect is with Ni Ci I ; ).· analog namely, compound 47 and OCH3 analog, compound 50. Reduced potency is seen with NHCOCH3, compound 49. This can be understood on the basis of OCII3 and N(CH3)2 being strong electron donors (Lewis bases and hydrogen bond donors) compared with the NHCO group with lower electron density on the nitrogen atom due to amide resonance.
[0115] In these studies, ibogaine, compound 47, and compound 50 were found to be more potent inhibitors than 18-MC (1 8-methoxycoronaridine) and noribogame on the epibatidine-induced calcium response in a3b4 nAChRs (left panel; DMX 1001 A is noribogame and OBI.215.21.1 is 18-MC). Compound 47 displayed a better affinity (5- fold) and selectivity (10-fold) for kOR than for mOR in comparison to 18-MC. In comparison to noribogaine, compound 47 gained affinity at kOR (X2), but mainly lost affinity to mOR resulting in a right-ward shift. Compounds 47 and 50 lost affinity for mOR and retained affinity for kOR. These results suggest that compounds 47 and 50 have improved selectivity while maintaining a3b4 activity.
Tabic 4: Evaluation of the relative potency of 18-MC and Noribogaine analogues (compounds 47-50) at a3b4 nAChRs
Figure imgf000035_0001
Example 7; nui and kappa opioid receptor binding activity
[0116) Noribogaine binds with high affinity (50 n ) to the 5-HT transporter and weak affinity at mu (900 nM) and kappa (1 μΜ) opioid receptors. At physiologically relevant concentrations, noribogaine only bound to SERT, DAT, mu and kappa opioid receptors (out of a panel of over 50 receptors, transporters, ion channels, second messengers, growth factor receptors and enzymes). The association between cigarette smoking and the presence and severity of suicidal behavior across major psychiatric disorders may be related to lower brain serotonin function in smokers with depression). Noribogainc analogs will likely retain affinity for the 5-HT transporter and we plan to fully characterize their activity in radioligand binding and functional uptake assays (serotonin and DA).
[01 17} Established radioligand binding methods are used to evaluate the ability of the compounds to block reuptake of serotonin and dopamine in membrane preparations of synaptosomes. Equilibrium binding affinity values is determined for each compound at opioid receptors (μ and κ subtypes) and biogenic amine transporters (DAT and SERT). Where possible, binding experiments utilize membranes derived from CHO or HEK293 cells engineered to overexpress the human form of the receptor or transporter of interest.
Radiolabeling of opioid receptors, and biogenic amine transporters utilizes conventional radioligands, buffers and incubation conditions according to published assays.
[0118] Test compounds, reference compounds and the radioligand ( d value) are added to binding buffer. Assays are run with test or reference compound over a range of concentration (0.0001 -10 uM) in triplicate using a 96-well sample plate containing radioligands specific for the target receptor or transporter. Cell membranes at the appropriate concentration are added in a volume of 400 μΐ in binding buffer. Following mixing on a plate shaker, samples are incubated for 1 -2 hours (depending on the transporter or receptor) at 25°C and filtered onto 96-well Unifilters (pre-treated with 0.3% polyefhyleneimine) using a Filtermate harvester. Bound radioactivity is counted in a Microbeta scintillation counter. Total bound radioactivity is estimated from triplicate wells containing no test or reference compound, together with nonspecific binding and filter blanks. The average bound radioactivity in the presence of the test compounds is expressed % inhibition = 100% - % radioactiv ity bound. The (( ';,, and i values for test and reference compounds are obtained by fitting normalized CPM data fitted using nonlinear (Graphpad Prism software).
Example 8: CNS penetration across the brain blood barrier
[01 19J Ibogaine, noribogaine, and 18-MC cross the BBB and reach micromolar concentrations in brain. Mice are dosed with compounds. Brain to blood ratios are determined by HPLC and EC/MS/MS analysis following oral administration of the test compounds in mice. Compound pharmacokinetic studies at representative time points are obtained in vivo in mice to demonstrate that the drug crosses the BBB following oral administrations, 5. Synthetic Methods
[0120] The compounds of this invention can be prepared from readily available starting materials using the following general methods and procedures. It will be appreciated that where typical or preferred process conditions (i.e., reaction temperatures, times, mole ratios of reactants, solvents, pressures, etc.) are given, other process conditions can also be used unless otherwise stated. Optimum reaction conditions may vary with the particular reactants or solvent used, but such conditions can be determined by one skilled in the art by routine optimi ation procedures.
[0121 J Additionally, as will be apparent to those skilled in the art, conventional protecting groups may be necessary to prevent certain functional groups from undergoing undesired reactions. Suitable protecting groups for various functional groups as well as suitable conditions for protecting and deprotecting particular functional groups are well known in the art. For example, numerous protecting groups are described in T. W. Greene and G. M. Wuts, Protecting Groups in Organic Synthesis, Third Edition, Wiley, New York, 1999, and references cited therein.
[0122] Furthermore, the compounds of this invention will typically contain one or more chiral centers. Accordingly, if desired, such compounds can be prepared or isolated as pure stereoisomers, i.e., as individual enantiomers or diastereomers, or as stereoisomer-enriched mixtures. All such stereoisomers (and enriched mixtures) are included within the scope of this invention, unless otherwise indicated. Pure stereoisomers (or enriched mixtures) may be prepared using, for example, optically active starting materials or stereoselective reagents well-known in the art. Alternatively, racemic mixtures of such compounds can be separated using, for example, chiral column chromatography, chiral resolving agents and the like.
[0123] Still further, some of the compounds defined herein include vinyl groups which can exist in cis, trans or a mixture of cis and trans forms. All combinations of these forms are within the scope of this invention.
|() 124] The starting materials for the following reactions are generally known compounds or can be prepared by known procedures or obvious modifications thereof. For example, many of the starting materials are available from commercial suppliers such as Aldrich Chemical Co. (Milwaukee, Wis., USA), Bach em (Torrance, Calif, USA), Emka-Chemce or Sigma (St. Louis, Mo., USA). Others may be prepared by procedures, or obvious modifications thereof, described in standard reference texts such as Fieser and Fieser's Reagents for Organic Synthesis, Volumes 1 15 (John Wiley and Sons, 1991 ), Rodd's Chemistry of Carbon Compounds, Volumes 1 5 and Supplcmcntals (Elsevier Science Publishers, 1989), Organic Reactions, Volumes 1 40 (John Wiley and Sons, 1 91 ), March's Advanced Organic Chemistry. (John Wiley and Sons, 4,sup.th Edition), and Larock's Comprehensive Organic Transformations (VCH Publishers Inc., 1989).
[0125] Compounds of this invention may be prepared using the starting material
Compound 1.1, which may be prepared according to Scheme 2, according to known procedures, such as those described in Bornmann, et al., ,/. Org. Chem. , 57: 1752 (1992) and U.S. Patent 6,21 1 ,360, which are hereby incorporated by reference in their entirety. Scheme 1 shows an exemplary general process for prepare compounds of this invention. The starting materials for the following reactions are generally known compounds or can be prepared by known procedures.
Scheme 1
Figure imgf000038_0001
[0126] Scheme 1 shows an exemplifying procedure to prepare compounds of Formula I, wherein R2, R3, L and R4 are as defined herein. Compound 1.1, wherein Pr1 is R1 as defined herein or a hydroxy protecting group, and Pr' is R as defined herein or a protecting group for the indole nitrogen, is converted to Compound 1.2 using the procedure described in uehne, et al. J. Org. Chem. 50:919 (1985). Hydroxyl protecting groups include a hydrolysable group as defined for Formula I, such as -C(0)R. wherein R is as defined herein, and other hydroxyl protecting groups known in the art, such as a benzyl (Bn) group, 2,4-dichlorobenzyl (DCB), methoxymethyl (MOM), tetrahydropyranyl (THP), acyl (such as acetyl (Ac)) or a silyl group, etc. Other suitable hydroxy protecting groups are known in the art. Protecting groups for the indole nitrogen include arylsulfonyl derivatives (e.g. tosyl (Ts)), carbamates (e.g.
fluorenylmethyloxycarbonyl (Fmoc) and tert-butyloxycarbonyl (t-BOC), trialkylsilyl groups (e.g. triisopropylsilyl). Ν,Ο-acetals (e.g. (trimethylsilyl)ethoxymethyl ether (SEM)) and certain substituted alkyl groups (e.g. benzyl). The need to protect and the types of protecting groups used for the hydroxy and/or the indole nitrogen will depend on the exact reaction conditions and reagents used in subsequent steps, which would be generally known in the art. When Pr1 is R1 and/or Pr2 is R2, they can be retained as part of the final compounds, or may be converted to other groups as defined in Formula I by methods generally known in the art. When Pr1 and/or Pr2 are other protecting groups, they can be removed under proper deprotection conditions to provide the free hydroxy group and/or free indole nitrogen, which may be further converted to the R1 and/or R2 groups as defined in Formula I.
[0127] Compound 1.2 may react with Compound 1.3 to give Compound 1.4 in dry toluene under reflux conditions, and preferably with a Dean-Stark trap with molecular sieves. Compound 1.3 can be prepared by using methods described in U.S. Patent 6,21 1 ,360, which is incorporated by reference in its entirety.
[0128] Alternatively, Compound 1.2 may react with Compound 1.3 to give a
condensation product in an organic solvent, such as alcohol solvents, such as methanol, ethanol, isopropanol, and n-butanol; ester-containing solvents, such as ethyl acetate and isopropyl acetate; ether solvents, such as tetrahydrofuran, diglyme, and dioxane; chlorinate hydrocarbons, such as methylene chloride, chloroform, and carbon tetrachloride; aromatic hydrocarbons, such as benzene, toluene, and xylene; acetonitrile: pyridine; and
dimethylformamide. The reaction may proceed under room temperature or under heated conditions, such refiexing conditions. After the condensation reaction, the condensation product is treated in a suitable solvent with an equivalent amount of an appropriate arylalkyl containing a good leaving group, such as an arylalkyl tosylate, an arylalkyl mesylate, or an arylalkyl halide, such as benzyl bromide, for 0.5 to 72 hours, for example, 16 hours, at 50 L C to 120 °C, which may be at the reflux temperature of the solvent. Suitable solvents include lower alkanes, such as pentane. hcxanc, or petroleum ether: aromatic hydrocarbon solvents, such as benzene, toluene, and xylene; alcohols, such as methanol, ethanol, isopropanol, and n-butanol; and ether solvents, such as diethyl ether, diglyme, or tetrahydrofuran.
[012 1 Treatment of the above product, with an organic-soluble Lewis base, such as tricthylaminc. produces a transient enamine acryiate of the formula:
Figure imgf000040_0001
[0130] This reaction can proceed in an organic solvent, including alcohol solvents, such as methanol, ethanol, isopropanol, and n-butanol; ketone solvents, such as acetone, methyl ethyl ketone, and cyclopentanone; ester-containing solvents, such as ethyl acetate and isopropyl acetate; ether solvents, such as tetrahydrofuran, diglyme, and dioxanc; chlorinated hydrocarbons, such as methylene chloride, chloroform, and carbon tetrachloride; acetonitrile; pyridine; and dimethylformamide. This reaction can be conducted at any temperature from room temperature to the boiling point of the solvent, for example, from 50 °C to 70 UC for, for example, from 1 to 10 hours. The transient enamine acryiate spontaneously cyclizes to give Compound 1.4.
Compound 1.4 undergoes reduction with a reducing agent, such as NaBH4, in the presence of acetic acid, under heating conditions, for example, at a temperature of between 80 °C and 1 10 °C, or between 85 °C and 95 °C, followed by catalytic hydrogenation in the presence of a catalyst, such as palladium, in a solvent, such as methanol, to provide Compound 1.5.
[0131] Hydrolysis of Compound 1.5 under acidic conditions, for example, with glacial acetic acid and/or hydrochloric acid in a solvent, such as MeOH gives Compound 1.6, which undergoes cyclization to give Compound 1.7 in a dry solvent under heating conditions, for example, in dry toluene at 130 °C.
|0132] Finally, Pr1 and/or Pf may be removed under suitable deprotection conditions to provide compounds where R1 and/or are hydrogen. One or both of these hydrogen atoms may be substituted by a desired R1 and/or R2 by reaction with R'Lg and/or R2Lg, wherein Lg is a leaving group, such as a halo group (e.g., chloro, bromo and iodo), sulfonate esters, such as para-toluenesulfonate or tosylate (OTs), hydroxy or alkoxy group. Further manipulations of the R1, R2, RJ and /or R4 to provide other embodiments of this invention are apparent to a person skilled in the art.
[0133] In Scheme 2, Compound 2.1 (5-hydroxyindole-3-acetic acid, which is
commercially available, for example, from Sigma- Aldrich Corp.) may be protected to give Compound 2.2, wherein Prl and Pr2 are as defined above. Compound 2.2 is reduced to the corresponding alcohol Compound 2.3, using a reducing agent, such as lithium
aluminumhydride. Reduction of Compound 2.2 to Compound 2.3 may optionally conducted via an intermediate aldehyde compound. The hydroxy group of Compound 2.3 is converted to a leaving group Lg2, such as a halo group (e.g., chloro. bromo and iodo), sulfonate esters, such as para-toluenesulfonate or tosylate (OTs), to give Compound 2.4. Compound 2.4 can react with ammonium to give Compound 1.1 or with a protected ammonium compound, such as NH(t-Boc)2 to give Compound 1.1 after deprotection of the protecting group. Other known methods of converting the acid group (-C(O)OH) to an methyl amino group (-CH2NH2) can also be used.
Scheme 2
Figure imgf000041_0001
[0134] By using commercially available 5-bromoindole-3-acetic acid and following
Scheme 2, the bromo compound corresponding to Compound 1.1 can be prepared, which can be used to replace Compound 1.1 in Scheme 1 to prepare compounds of this invention wherein R is bromo. The bromo atom can be converted to other R groups as defined for f ormula 1 using procedures known in the art. For example, reaction with a suitable agent, such as haloalkyk haloalkenyl, or haloalkynyl, optionally in the presence of a copper (I) or palladium catalyst and/or a base, such as triethylamine, would give compounds where R is alkyl (when using haloalkenyl or haloalkynyl, the double bond or triple bond can be reduced by hydrogcnation).
|0135| Scheme 3 shows an example of preparing an intermediate Compound 3.5, which may be used in Scheme 1 to replace Compound 1.1 to prepare compounds of this invention where R is chloro. In Scheme 3. Compound 3.1 (5-chloroindole-3-carboxaldehyde, which is commercially available, for example, from Sigma- Aldrich Corp.) is protected with Pr2 to give Compound 3.2. Compound 3.2 may react with a Wittig agent, e.g. PhsPCff^B . to give Compound 3.3. Compound 3.3 is converted to Compound 3.4, wherein Halo is a halo such as Br or CI. by reacting with, for example, hydrogen bromide (HBr). Compound 3.4 can react with ammonium to give Compound 3.5 or with a protected ammonium compound, such as NH(t-Boc)2 to give Compound 3.5 after deprotection of the protecting group. Other known methods of converting the acid group (-C(O)OH) to an methyl amino group (- CH2NH2) can also be used.
Scheme 3
Figure imgf000042_0001
[0136] Further compounds of the present invention can be prepared according to Scheme 4 from noribogaine derivatives (as can be prepared by the synthesis disclosed above, for example) by methods known to one of skill in the art. Λ skilled artisan would appreciate that the reactivity of the hydroxy group and the indole nitrogen is different so that selectivity can be achieved by selecting suitable reagents and suitable reaction conditions for one of them to react but leaving the other intact to form the desired product. For example, the hydroxy group is expected to selectively react with a carboxylic acid in the presence of
triphenylphosphine (P1¾P) and diethyl azodicarboxylate (DEAD) to give Compound 4.1 and Compound 4.3 where L is -C(0)alkylene or -C(0)arylene. Thus, as shown in Scheme 4, noribogaine derivatives can react with appropriately protected compounds of formula LG-R30 where R30 is -L ' -R! 7. -L! -R I S, -L '-R19 or -L' -CHR17R, s, LG is a leaving group such as hydroxy, alkoxy, halo, etc., to give Compound 4.2, which may further react with LG-R12 to form Compound 4.3, In other embodiments (i.e. when R is H), the C12-phenol, thiol, or amino group is protected by reaction with a suitable protecting group, PG-LG. where LG is a leaving group such as defined above, such that the indole nitrogen is selectively derivatized with R12. Suitable protecting groups are well kno B in the art (see T. W. Greene, P. G. M. Wuis, Protective Groups in Organic Synthesis, 4th Edition, Wiley-Interscience, New York, 2006). In another alternative embodiment (i.e. when R12 is H). the indole nitrogen is protected with a suitable protecting group, PG (see Greene et al., supra), such that the C I 2- phenol, thiol, or amino group is free to be derivatized with R^°.
Scheme 4
Figure imgf000043_0001
Alternatively, as shown in Scheme 5, certain compounds of this invention may be prepared by reacting ibogaine with LG-R12 to give Compound 5.1. Compound 5.1 can be demethylated by methods known in the art, such as reaction with boron tribromide/methylene chloride at room temperature to give Compound 5,2, which may further react with LG~R~'° to give Compound 5.3, Scheme 5
Figure imgf000044_0001
5.4
[0137] Compounds of formula IC are prepared from a known starting material, voacangine, as shown in the scheme 6 below.
Scheme 6
Figure imgf000044_0002
6.3
[01 81 The carboxymethyl group in Voacangine is hydrolyzed or converted into litium salt using an organolithium reagent such as butyllithium and proapancthiol in a solvent such as HMl'A/ I HF. The lithium salt is then converted to the acid chloride 6,2 by reacting with oxalyl chloride in presence of a base such as pyridine in a solvent such as THF. The acid chloride 6.2 is then esterfied with an alcohol to give the ester 6.3. Demethylalion of the ester with a reagent such as BBr;, in a solvent such as methylene chloride gives compounds of formula IC. J0139] Alternatively, as shown in Schemes 7 and 8, certain compounds of this invention may be prepared using noribogaine, which may be prepared according to known procedures, such as by demethylating ibogaine by methods known in the art, such as reaction with boron tribromi de/methy 1 ene chloride at room temperature.
[01401 Schemes 7 and 8 below show reaction schemes for the sulfation of the 12 -hydroxy 1 group and optionally for the disulfation of the 12-hydroxyl group and the indole nitrogen atom. Scheme 8 below shows reaction schemes for selective sulfation of the indole nitrogen atom by protecting the 12-hydroxyl group with a conventional hydroxyl protecting group. A variety of protecting groups, preferably those stable under acidic conditions are useful as the Pg, as will be apparent to the skilled artisan. An ester of the chlorosulfonic acid may be used to prepare an ester of the compound of Formula I. It is also contemplated that the indole nitrogen of noribogaine can be protected, the sulfation carried out on the hydroxy group of noribogaine, following which, the N-protecting group is deprotected. Methods for preparing the N-protected noribogaine will be apparent to the skilled artisan in view of this disclosure.
Scheme 7
Figure imgf000045_0001
7.1 7.2
Scheme 8
Figure imgf000045_0002
[0141] As shown above, X refers to a leaving group such a chloro, bromo, iodo, or a R5- S03-moiety, where Rs ia Ci-C6 alkyl optionally sublimed with 1 -3 fluoro atoms or Rs is phenyl optionally substituted with 1-3 halo or Ci-C6 alkyl groups.
[0142] The dihydronoribogaine compounds of Formula IB are synthesized by reducing the double bond of the corresponding noribogaine derivative. Various reducing agents well known to the skilled artisan are useful for this purpose. For example, cataly tic hydrogenalion employing hydrogen and a catalyst such as Pd/C or Pl/C is useful for providing the 9, 17 as. i.e. the a,a or the β,β dihydro compounds. Reagents such as borohydride or aluminum hydrides are useful for providing the α,β or the β,α dihydro compounds.
[0143] As shown in Schemes 7 and 8, the phosphate derivatives at 12-hydroxy group, indole nitrogen, and both positions are synthesized in an analogous manner via
phosphorylation instead of sulfation.
[0144] Compounds of this invention as represented by Formula 1 -1 can be prepared from noribogaine using an appropriate phosphate source, such as phosphoric acid or a
phosphoramidite such as di-fcr/-butyl N,N-diisopropylphosphoramidite. Compounds of Formula 1-2 can be prepared from compounds of Formula 1-1 using an appropriate phosphate source under known reaction conditions. The reactions are carried out for a period of time sufficient to provide a substantial amount of the product, which can be ascertained by- using routine methods such as thin layer chromatography, Ή-nuclear magnetic resonance (NMR) spectroscopy, and the likes. Compounds of Formula 1-1 and 1-2 can be isolated and optionally purified using standard purification techniques, such as liquid chromatography. Scheme 2 follows much of the chemistry of Scheme 1 with the exception that a blocking (protecting group - Pg) is used to avoid phosphorylation of the 12 hydroxyl group.
[0145] The dihydronoribogaine compounds of Formulas 1, 1-A, and 11 are synthesized by reducing the corresponding double bond of noribogaine. Various reducing agents well known to the skilled artisan are useful for this purpose. For example, catalytic hydrogenalion employing hydrogen and a catalyst such as Pd/C or Pt/C is useful for providing the 9, 1 cis, i.e. the α,α or the β,β dihydro compounds. Reagents such as borohydride or aluminum hydrides are useful for providing the α,β or the β,α dihydro compounds.
[0146] It will be apparent to those skilled in the art that many modifications of the above exemplifying methods, both to materials and methods, may be practiced without departing from the scope of the c urrent invention.

Claims

What is claimed is:
1. A compound Formula IA or Formula IB, or a pharmaceutically acceptable salt of each thereof:
Figure imgf000047_0001
IA IB
wherein
R is OR1 or Ci-Cn alkyl optionally substituted with 1 to 5 R9;
R1 is selected from the group consisting of hydrogen, -C(0)OX, -S02ORH), a
monophosphate, a diphosphate, a triphosphate, and -C(0)N(Y)2 where X is C| -C6 alkyl optionally substituted with 1 to 5 R9, and each Y is independently selected from the group consisting of hydrogen, tVG, alkyl optionally substituted with 1 to 5 R9, C -C 14 aryl optionally substituted with 1 to 5 R9, C3-C10 cycloalkyl optionally substituted with 1 to 5 R9, Ci -Cio heteroaryl having 1 to 4 heteroatoms and which is optionally substituted with 1 to 5 R9, C|-C 10 heterocyclic having 1 to 4 heteroatoms and which is optionally substituted with 1 to 5 R9, or where each Y, together with the nitrogen atom bound thereto form either a C i -C(, heterocyclic having 1 to 4 heteroatoms and which is optionally substituted with 1 to 5 R9, or a Ci -Ce heteroaryl having 1 to 4 heteroatoms and which is optionally substituted with 1 to 5 Ry;
R is halo, OR20, or C-1 -C12 alkyl optionally substituted with 1 to 5 R9;
R" is selected from the group consisting of hydrogen , -C(0)X, -C(0)OX and -C(0)N(Y)2 where X and Y are defined as above;
R2 is hydrogen, -SOiOR10, a monophosphate, a diphosphate, or a triphosphate;
R3 is selected from the group consisting of hydrogen, -(CEhjmOR7, -C 6(OH)R', -(CH2)mCN, -(CH2)mCOR7, -(CH2)mC02R7, -(CH2)mC(0)NR6R7, -(CH2)mC(0)NR NR7R7, -(CH2)mC(0)NR6NR7C(0)R8, and -(CH2)mNR6R7;
m is 0, 1 , or 2; L is a bond or Cj-Ci? alkylene;
R4 is selected from the group consisting of C C 12 alkyl substituted with 1 to 5 R9, C2-Q2 alkenyl substituted with 1 to 5 R". -X2-R6, -( X2-Y2)„-X -R6, -S02NR6R \ -0-C(0)R8, - (0)OR7, -C'lOjNR'R7, -NR6R7. -NHCfO)R8, and -NR6C(0)R8;
X2 is selected from the group consisting of O and S;
Y2 is C1 -C4 alkylene or C6-C10 arylene, or a combination thereof;
n is 1 , 2, or 3;
R6 and R7 are each independently selected from the group consisting of hydrogen, Cj -C alky] optionally substituted with 1 to 5 R9, C | -C6 heterocycle having 1 to 4 heteroatoms which is optionally substituted with 1 to 5 R9, CYCi o cycloalkyl optionally substituted with 1 to 5 R\ C6-C|o aryl optionally substituted with 1 to 5 R9 and C 1 -C6 heteroaryl having 1 to 4 heteroatoms optionally substituted with 1 to 5 R9; or
R6 and R are joined to form Q-CG heterocycle having 1 to 4 heleroatoms which is optionally substituted with 1 to 5 R9;
R8 is selected from the group consisting of C 1 -C 12 alkyl optionally substituted with 1 to 5 R9, C] -C(, heterocycle having 1 to 4 heteroatoms optionally substituted with 1 to 5 R9, C3- Cio cycloalkyl optionally substituted with 1 to 5 R9, C6-Cio aryl optionally substituted with 1 to 5 R9 and C \ -C(, heteroaryl having 1 to 4 heteroatoms optionally substituted with 1 to 5 R9;
R9 is selected from the group consisting of C 1 -C4 alkyl, phenyl, halo, -OH, -OR10, -CN, -COR10, -CO2R1 , -C(0)NHR1 0, -NRI 0R10, -C(O)NR, 0RU), -C(0)NHNHRKJ, -C(0)NR l uNHRl <J, -C(O)NR10NR10R10, -C(O)NHNR10C(O)R1 ,
-C(0)NHNHC(0)R10, -S02NR1(,R10, -C(O)NR, 0NRL 0C(O)R,°, -NHC(0)R10, C,-C6 heterocycle having 1 to 4 heteroatoms optionally substituted with 1 to 5 R10, and -C(O)NR!0NHC(O)R10; and
R! 0 is CrC i, alkyl;
provided that:
for the compound of Formula IA, when R is -OH or C1-C12 alkyl optionally substituted with 1 to 5 R9, then R~ is hydrogen; and for the compound of Formula Ι.Λ. when R is hydrogen, and -L-R4 is ethyl, then R is not
-OR'.
2. A compound of Formula IIA or Formula IIB, or a pharmaceutically acceptable salt of each thereof:
Figure imgf000049_0001
wherein
wherein
R" is halo, -OH, -SH, -NH2, -S(0)2N(R16)2, -X'-L'-R17, -X'-L'-R18, -X'-L'-R19, or- X'-L'-CHR1^18, where X1 is O, S or NR16, or -O-Q, where Q is -S02-(Ci-CL2 alkyl), a monophosphate, a diphosphate, or a triphosphate;
L1 is alkylene, arylene, -C(0)-alkylene, -C(0)-arylene, -C(0)0-arylene, -C(0)0- alkyiene, -C(0)NR19-alkyiene, -C(0)NR19-arylene, -C(NR19)NR19-alkylene or
-C(NR19)NRiy-arylene, wherein L1 is configured such that -O-L'-R1 ' is - OC(0)-alkylene-R17, -OC(0)0-arylene-R17, -OC(0)0-alkylene-R17, -OC(O)- arylene-R17, -OC(0)NRl9-alkylene-R17, -OC(0)NRl9-arylene-R17, - OC(NR19)NRl9-alkylene-R17 or -OC( iRl9)NR19-arylene-R17, and wherein the alkylene and arylene are optionally substituted with 1 to 2 R ;
R12 is hydrogen, -S(0)20R19, -S(0)2R19, -C(0)R14, -C(0)NRl4R14, -C(0)OR14, C Cu alkyl optionally substituted with 1 to 5 R]\ Ci-Ci2 alkenyl optionally substituted with 1 to 5 Rls, aryl optionally substituted with 1 to 5 R , -S02- (C1-C12 alkyl), a monophosphate, a diphosphate, or a triphosphate;
R'3 is hydrogen, halo, -OR16, -CN, C1-C12 alkyl, C 1 -C 12 alkoxv, aryl or aryloxy, where the alkyl, alkoxy, aryl, and aryloxy are optionally substituted with 1 to 5 Rls; each R14 is independently selected from the group consisting of hydrogen, C1-C12 alkyl, C2-Ci2 alkenyl, C2-Ci2alkynyl, aryl, heteroaryl. and heterocycle, and wherein the alkyl, alkenyl. alkynyl, aryl, heteroaryl, and heterocycle are optionally substituted with 1 to 5 R15;
R° is selected from the group consisting of phenyl, halo, -OR16, -CN, -COR16,
-C02R16, -NR16R16, - NR,6C(0)R16, - NR16S02R16, -C(0)NRI6R16,
-C(()) R,6NRI6R16, -S02NR16R16 and -C(0) RI6 RI6C(0)R16; each R16 is independently hydrogen or C]-C 12 alkyl optionally substituted with from 1 to 3 halo;
R1 is hydrogen, -C(0)R19, -C(0)OR19, -C(0)N(R19)2 or -N(R19)C(0)R19;
RIS is hydrogen, -N(R,9)2, -C(0)N(R19)25 -C( R1 )N(R!9)2, -C(NS02RI9)N(R,9)25 -NR19C(0)N(RL9)2J -NR,9C(S)N(R19)2, - RI9C(NRI9)N(R19)2, -NR,9C(NS02R19)N(R19)2 or tetrazole; and
each R19 is independently selected from the group consisting of hydrogen, ( .-( . ^ alkyl and aryl;
provided that:
for the compound of Formula ΠΑ:
when R12 and R13 are hydrogen, then R11 is not hydroxy;
when R13 is hydrogen, R is -O-L'-R17, -O-L'-R18, -O-L'-R19, and L1 is
alkylene, then -O-L'-R17, -O-L'-R18, -O-L'-R19 are not methoxy; and when R13 is hydrogen, X1 is O, L is -C(0)-alkylene, -C(0)-arylene, -C(0)0- arylene, -C(0)0-alkylene, -C(0)NR,9-alkylene, or -C(0)NRI9-arylene, then none of R17, R18 or R19 are hydrogen.
3, The compound of claim 2, represented by Formula IJC or Formula IID, or a pharmaceutically acceptable salt of each thereof:
Figure imgf000050_0001
lie IID
4. The compound of claim 2, wherein R is halo.
5. The compound of claim 2, wherein R is bromo.
6. The compound of claim 2, wherein R1 1 is -OH, -O-L'-R1 ', -O-L'-R1 8, -O-L' -R19 or - 0-L'-CHR, 7R, R.
7. The compound of claim 2, wherein R1 1 is -SH, -S-lJ-R17, -S-L'-R18, -S-L' -R 19 or -S-
8. The compound of claim 2, wherein R1 1 is NH2, -NRI 6-L' -R1 7, -NR^-L'-R18, -NR16- L'-R^ or -NR^-L' -CHR' 8.
9. The compound of claim 2, wherein R1 1 is -S(0)2N(R16)2.
10. The compound of claim 2, wherein R1 1 is -CI, -Br, -1, -OC(0)CH2CH3, - OC(0)CH2Ph, -OC(0)OCH2CH3, -OC(0)OCH2Ph, -OC(0)NH(CH(CH3)Ph), - OC(NPh)NHCH2CH3, -OC(0)NHCH2Ph, -NH2, -NHC(0)C(CH3)3; -NHC(0)CF3,
-NHC(0)CH2Ph, -NHC(0)OCH2Ph, -NHC(0)NH(CH(CH3)Ph), -SH , -SC(0)OCH2Ph, -SC(0)NH(CH(CH3)Ph), -SC(0)CH2CH2OP(0)(OH)2; -OC(0)CH2CH2CH(NH2)(C02H), - 0-(2-OH-C6H3)CH2CH(NH2)(C02H) or -NHC(0)CH2CH2CH(NH2)(C02H).
1 1. The compound of claim 2. wherein R12 is hydrogen.
12. The compound of claim 2, wherein R12 is -CH2CH3, -CH2CHCH2, -CH2Ph,
-C(0)0(CI I2)2N(CH3)2 or -C(0)CH2(CH2)2S02N(CH3)2.
13. The compound of claim 2, wherein R13 is hydrogen.
14. The compound of claim 2, wherein R13 is alkyl substituted with from 1 to 3 halo.
15. The compound of claim 2, wherein R is trifluoromethyl.
16. The compound of claim 2, wherein RN is ^OCH3, -OCH2CH3, -OCH2Ph or ^CN.
1 7. A compound of Formula i!E or a pharmaceutically acceptable salt thereof;
Figure imgf000051_0001
HE wherein the compound is selected form the group consisting of compounds wherein R , R RL' are defined as follows:
Figure imgf000052_0001
Figure imgf000053_0001
-NHC(0)C(CH3)3 H H single bond
Figure imgf000054_0001
46 bond
18. A compound of formula IC :
Figure imgf000054_0002
I C
or a pharmaceutically acceptable salt thereof, wherein the compound is selected form the group consisting of compounds wherein R10 is defined as follows:
Table 2
Figure imgf000055_0001
19. A composition comprising a compound of claim 1 or 2 and a pharmaceutically acceptable excipient.
20. A method of treating pain and/or addiction in a patient, comprising administering to said patient a compound of claim 1 or 2 or a pharmaceutically acceptable salt thereof or a composition of claim 19.
21 . The method of claim 20, which method is for treating neuropathic pain.
22. The method of claim 20, which method is for treating nociceptive pain.
23. The method of claim 20, which method alleviates a symptom associated with withdrawal from drag dependency selected from the group consisting of nausea, vomiting, anxiety, abdominal cramps, muscle pain, chills and headache.
24. The method of claim 20. wherein the addiction is an addicted to ***e, alcohol, amphetamines or combinations thereof.
25. The method of claim 20, further comprising administering to the patient an opioid antagonist selected from the group consisting of naloxone, naltrexone and nalorphine.
26. A method of treating nicotine addiction in a patient comprising administering to said patient an effective amount of a compound of claim 1 or 2 or a pharmaceutically acceptable sail thereof or a composition of claim 19.
27. A method of inhibiting a3 b4 nicotinic acetylcholine receptors comprising administering an effective amount of a compound of claim 1 or 2 or a pharmaceutically acceptable salt thereof or a composition of claim 19.
PCT/US2012/071052 2012-12-20 2012-12-20 Substituted noribogaine WO2014098877A1 (en)

Priority Applications (8)

Application Number Priority Date Filing Date Title
CA2896133A CA2896133A1 (en) 2012-12-20 2012-12-20 Substituted noribogaine
JP2015549336A JP2016508979A (en) 2012-12-20 2012-12-20 Substituted norbogaine
US14/654,503 US9783535B2 (en) 2012-12-20 2012-12-20 Substituted noribogaine
PCT/US2012/071052 WO2014098877A1 (en) 2012-12-20 2012-12-20 Substituted noribogaine
EP12890228.5A EP2934541A4 (en) 2012-12-20 2012-12-20 Substituted noribogaine
US13/732,751 US8940728B2 (en) 2012-12-20 2013-01-02 Substituted noribogaine
US13/753,364 US9045481B2 (en) 2012-12-20 2013-01-29 Substituted noribogaine
US14/719,041 US20150361080A1 (en) 2012-12-20 2015-05-21 Substituted noribogaine

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
PCT/US2012/071052 WO2014098877A1 (en) 2012-12-20 2012-12-20 Substituted noribogaine

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US13/732,751 Continuation US8940728B2 (en) 2012-12-20 2013-01-02 Substituted noribogaine
US13/753,364 Continuation US9045481B2 (en) 2012-12-20 2013-01-29 Substituted noribogaine

Publications (1)

Publication Number Publication Date
WO2014098877A1 true WO2014098877A1 (en) 2014-06-26

Family

ID=50978949

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2012/071052 WO2014098877A1 (en) 2012-12-20 2012-12-20 Substituted noribogaine

Country Status (5)

Country Link
US (1) US9783535B2 (en)
EP (1) EP2934541A4 (en)
JP (1) JP2016508979A (en)
CA (1) CA2896133A1 (en)
WO (1) WO2014098877A1 (en)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2018210229A1 (en) * 2017-05-18 2018-11-22 南京明德新药研发股份有限公司 α-7 NICOTINE ACETYLCHOLINE RECEPTOR AGONIST AND APPLICATION THEREOF
US10519175B2 (en) 2017-10-09 2019-12-31 Compass Pathways Limited Preparation of psilocybin, different polymorphic forms, intermediates, formulations and their use
US11564935B2 (en) 2019-04-17 2023-01-31 Compass Pathfinder Limited Method for treating anxiety disorders, headache disorders, and eating disorders with psilocybin

Families Citing this family (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9586954B2 (en) 2010-06-22 2017-03-07 Demerx, Inc. N-substituted noribogaine prodrugs
WO2013085922A1 (en) * 2011-12-09 2013-06-13 Demerx, Inc. Phosphate esters of noribogaine
WO2014098877A1 (en) * 2012-12-20 2014-06-26 Demerx, Inc. Substituted noribogaine
US9045481B2 (en) * 2012-12-20 2015-06-02 Demerx, Inc. Substituted noribogaine
WO2020181194A1 (en) * 2019-03-07 2020-09-10 University Of Padova Compositions and methods of use comprising substances with neural plasticity actions administered at non-psychedelic/psychotomimetic dosages and formulations

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3557126A (en) * 1969-08-14 1971-01-19 American Home Prod Derivatives of iboga alkaloids
US6211360B1 (en) * 1995-08-08 2001-04-03 Albany Medical College Ibogamine congeners
US6348456B1 (en) * 1994-07-25 2002-02-19 Deborah C. Mash Method of treating chemical dependency in mammals and a composition therefor
US20030158202A1 (en) * 2001-06-11 2003-08-21 Patrizia Caldirola New compounds
US8362007B1 (en) * 2010-05-11 2013-01-29 Demerx, Inc. Substituted noribogaine

Family Cites Families (98)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US2877229A (en) * 1959-03-10 Process for the manufacture of indoles
US2813873A (en) 1957-11-19 Derivatives of the ibogaine alkaloids
GB841697A (en) 1956-03-22 1960-07-20 Ciba Ltd Analgesic compositions
GB924042A (en) 1959-03-05 1963-04-18 Geigy Ag J R Process for the decarboxylation of alkyl esters of unsaturated, nitrogen-containing carboxylic acids
US3639408A (en) 1967-06-03 1972-02-01 Shionogi & Co Process for the preparation of isoquinuclidine derivatives
US3516989A (en) 1967-10-02 1970-06-23 American Home Prod Intermediates for total synthesis of iboga alkaloids and means of preparation
FR7131M (en) 1968-01-31 1969-07-21
US3574220A (en) 1969-08-14 1971-04-06 American Home Prod 1,7-methano-1h-1-benzazepin-5(4h)-ones
CA982478A (en) 1971-04-12 1976-01-27 Jerome Berk Orally administered drug composition for therapy in the treatment of narcotic drug addiction
US4375414A (en) 1971-05-20 1983-03-01 Meir Strahilevitz Immunological methods for removing species from the blood circulatory system and devices therefor
US3715361A (en) 1971-10-08 1973-02-06 American Cyanamid Co Acyl derivatives of 10-methoxyibogamine
US3875011A (en) 1972-11-06 1975-04-01 Syva Co Enzyme immunoassays with glucose-6-phosphate dehydrogenase
BE796521A (en) * 1973-03-09 1973-07-02 Omnium Chimique Sa N-substd indole derivs prepn - esp pharmaceutically active alkaloids
US4107288A (en) 1974-09-18 1978-08-15 Pharmaceutical Society Of Victoria Injectable compositions, nanoparticles useful therein, and process of manufacturing same
FR2296418B1 (en) 1974-12-30 1978-07-21 Anvar
US4272541A (en) 1978-06-02 1981-06-09 Miles Laboratories, Inc. 7,8 and 7-8 Substituted 4,5α-epoxymorphinan-6-one compounds, and methods of treating pain and drug dependence with them
AU7821581A (en) 1980-12-17 1982-06-24 Sterling Drug Inc. Substitution of aromatic organic compounds
US4464378A (en) 1981-04-28 1984-08-07 University Of Kentucky Research Foundation Method of administering narcotic antagonists and analgesics and novel dosage forms containing same
CA1188989A (en) 1981-05-21 1985-06-18 Richard R. Scherschlicht Nonapeptide for the treatment of drug withdrawal symptoms
US4604365A (en) 1981-06-02 1986-08-05 Electro-Nucleonics, Inc. Immunoprecipitation assay
US4462941A (en) 1982-06-10 1984-07-31 The Regents Of The University Of California Dynorphin amide analogs
US4499096A (en) 1983-11-18 1985-02-12 Lotsof Howard S Rapid method for interrupting the narcotic addiction syndrome
US5266574A (en) 1984-04-09 1993-11-30 Ian S. Zagon Growth regulation and related applications of opioid antagonists
US4626539A (en) 1984-08-10 1986-12-02 E. I. Dupont De Nemours And Company Trandermal delivery of opioids
US4573995A (en) 1984-10-09 1986-03-04 Alza Corporation Transdermal therapeutic systems for the administration of naloxone, naltrexone and nalbuphine
GB8430346D0 (en) 1984-11-30 1985-01-09 Reckitt & Colmann Prod Ltd Analgesic compositions
ATE95430T1 (en) 1984-12-22 1993-10-15 Sanol Arznei Schwarz Gmbh ACTIVE PATCHES.
US4806341A (en) 1985-02-25 1989-02-21 Rutgers, The State University Of New Jersey Transdermal absorption dosage unit for narcotic analgesics and antagonists and process for administration
US4587243A (en) 1985-07-15 1986-05-06 Lotsof Howard S Rapid method for interrupting the ***e and amphetamine abuse syndrome
US5316759A (en) 1986-03-17 1994-05-31 Robert J. Schaap Agonist-antagonist combination to reduce the use of nicotine and other drugs
US4904768A (en) 1987-08-04 1990-02-27 Bristol-Myers Company Epipodophyllotoxin glucoside 4'-phosphate derivatives
US4857523A (en) 1988-07-18 1989-08-15 Nda International, Inc. Rapid method for attenuating the alcohol dependency syndrome
US5629307A (en) 1989-10-20 1997-05-13 Olney; John W. Use of ibogaine in reducing excitotoxic brain damage
US5075341A (en) 1989-12-01 1991-12-24 The Mclean Hospital Corporation Treatment for ***e abuse
DE4010079A1 (en) 1990-03-29 1991-10-02 Lohmann Therapie Syst Lts PHARMACEUTICAL FORMULATION FOR THE TREATMENT OF ALCOHOLISM
US5026697A (en) 1990-05-30 1991-06-25 Nda International, Inc. Rapid method for interrupting or attenuating the nicotine/tobacco dependency syndrome
US5152994A (en) 1990-05-31 1992-10-06 Lotsof Howard S Rapid method for interrupting or attenuating poly-drug dependency syndromes
US5145684A (en) 1991-01-25 1992-09-08 Sterling Drug Inc. Surface modified drug nanoparticles
US5283247A (en) 1991-02-06 1994-02-01 Research Corporation Technologies, Inc. Anticonvulsant substituted quinazolones
US5149538A (en) 1991-06-14 1992-09-22 Warner-Lambert Company Misuse-resistive transdermal opioid dosage form
US5290784A (en) 1991-07-18 1994-03-01 Yueqian Qu Aconitane derivatives used as a medication to treat addiction
JP3713668B2 (en) 1992-04-10 2005-11-09 メレルファーマスーティカルズ インコーポレイテッド Cancer treatment agent by co-therapy of 2'-halomethylidene derivative and antineoplastic agent specific to S phase or M phase
JPH08501075A (en) 1992-06-12 1996-02-06 ディー イー エス − ティー ワイ アール ダイノルフィン パートナーシップ des-Tyr dynorphin analogue
US5256669A (en) 1992-08-07 1993-10-26 Aminotek Sciences, Inc. Methods and compositions for treating acute or chronic pain and drug addiction
US5382657A (en) 1992-08-26 1995-01-17 Hoffmann-La Roche Inc. Peg-interferon conjugates
WO1994006426A1 (en) 1992-09-21 1994-03-31 Qin Bo Yi Methods for identifying and using low/non-addictive opioid analgesics
US5580876A (en) 1992-09-21 1996-12-03 Albert Einstein College Of Medicine Of Yeshiva University, A Division Of Yeshiva University Method of simultaneously enhancing analgesic potency and attenuating dependence liability caused by morphine and other bimodally-acting opioid agonists
CN1042895C (en) 1992-09-23 1999-04-14 曲曰谦 Medicines composition and its method for treating drop syndrome and de-addiction of drugaddict
WO1994014490A1 (en) 1992-12-23 1994-07-07 Bernhard Hugemann Compacted drug body for use in the mechanical generation of inhalable active-substance particles
US5865444A (en) 1994-03-21 1999-02-02 Deere & Company Body leveling suspension including a pivoting arrangement
JP2819236B2 (en) 1994-05-06 1998-10-30 日東電工株式会社 Transdermal formulation
US5552406A (en) 1994-06-17 1996-09-03 The Mclean Hospital Corporation Method for treating pain and brain perfusion abnormalities using mixed opioid agonist-antagonists
US5591738A (en) 1994-10-14 1997-01-07 Nda International, Inc. Method of treating chemical dependency using β-carboline alkaloids, derivatives and salts thereof
US5616575A (en) 1995-12-04 1997-04-01 Regents Of The University Of Minnesota Bioactive tricyclic ibogaine analogs
US5760044A (en) 1996-05-16 1998-06-02 Archer; Sydney Method for treating ***e and amphetamine dependency
WO1997044317A2 (en) 1996-05-21 1997-11-27 The United States Of America, Represented By The Secretary, Department Of Health And Human Services Novel methods of o-demethylation and n-deprotection
GB9702701D0 (en) 1997-02-01 1997-04-02 Univ Newcastle Ventures Ltd Quinazolinone compounds
AU754088B2 (en) 1997-09-04 2002-11-07 Demerx, Inc. Noribogaine in the treatment of pain and drug addiction
US7220737B1 (en) 1997-09-04 2007-05-22 Novoneuron, Inc Noribogaine in the treatment of pain and drug addiction
ITTO980264A1 (en) * 1998-03-26 1999-09-26 Silvio Rossi APPLICATION OF A NON-QUATERNARY BASIC EXTRACT OF FUCHSIAEFOLIA PESCHIERA WITH ANTI-MALARICA ACTIVITY
US6451806B2 (en) 1999-09-29 2002-09-17 Adolor Corporation Methods and compositions involving opioids and antagonists thereof
US20030153552A1 (en) 2002-02-14 2003-08-14 Mash Deborah C. Method of treating chemical dependency in mammals and a composition therefor
US20030199496A1 (en) * 2002-04-22 2003-10-23 Simon David Lew Pharmaceutical compositions containing alpha3beta4 nicotinic receptor antagonists and methods of their use
US6864271B2 (en) 2002-11-12 2005-03-08 The Foundation For The Lsu Health Sciences Center Synergistic combinations including N-acylated 4-hydroxyphenylamine derivatives
EP1491197A1 (en) * 2003-06-26 2004-12-29 Bertelli Motta, Guiseppe Pharmaceutical composition comprising voacamine for the stimulation and modulation of the human immunologic system
US6806291B1 (en) 2003-10-09 2004-10-19 The Foundation For The Lsu Health Sciences Center Analgesic compounds, their synthesis and pharmaceutical compositions containing them
US20070244075A1 (en) 2003-11-18 2007-10-18 Novogen Research Pty Ltd Isoflavonoid Prodrugs, Compositons Thereof and Therapeutic Methods Involving Same
US8017151B2 (en) 2004-09-07 2011-09-13 Board Of Regents Of The University Of Nebraska By And Behalf Of The University Of Nebraska Medical Center Amphiphilic polymer-protein conjugates and methods of use thereof
AU2006274197A1 (en) 2005-07-26 2007-02-01 Merckle Gmbh Macrolide conjugates of pyrrolizine and indolizine compounds as inhibitors of 5-lipooxygenase and cyclooxygenase
DE602006018565D1 (en) 2005-08-26 2011-01-13 Medical Res Council London Anionically substituted 7-nitroindole derivatives and their use
CN101316840B (en) 2005-11-30 2012-08-08 霍夫曼-拉罗奇有限公司 1,5-substituted indol-2-yl amide derivatives
US7960544B2 (en) 2005-12-16 2011-06-14 Ironwood Pharmaceuticals, Inc. Useful indole compounds
CN1915993A (en) * 2006-09-12 2007-02-21 中国人民解放军第二军医大学 Indole group alkaloid of Hainan Gouyahua, and application for preparing drug-breaking medicine
US9625475B2 (en) 2008-09-29 2017-04-18 Abbvie Inc. Indole and indoline derivatives and methods of use thereof
JP2012504137A (en) 2008-09-29 2012-02-16 アボット・ラボラトリーズ Indole derivatives and indoline derivatives and methods for their use
CN106822083A (en) 2009-08-27 2017-06-13 生物学特性有限公司 Conjoint therapy for treating proliferative disease
US8765737B1 (en) 2010-05-11 2014-07-01 Demerx, Inc. Methods and compositions for preparing and purifying noribogaine
US9394294B2 (en) 2010-05-11 2016-07-19 Demerx, Inc. Methods and compositions for preparing and purifying noribogaine
US8741891B1 (en) * 2010-06-22 2014-06-03 Demerx, Inc. N-substituted noribogaine prodrugs
US8637648B1 (en) 2010-06-22 2014-01-28 Demerx, Inc. Compositions comprising noribogaine and an excipient to facilitate transport across the blood brain barrier
US9586954B2 (en) 2010-06-22 2017-03-07 Demerx, Inc. N-substituted noribogaine prodrugs
US8802832B2 (en) 2010-06-22 2014-08-12 Demerx, Inc. Compositions comprising noribogaine and an excipient to facilitate transport across the blood brain barrier
CA2806232A1 (en) 2010-07-23 2012-01-26 Demerx, Inc. Noribogaine compositions
EP2481740B1 (en) 2011-01-26 2015-11-04 DemeRx, Inc. Methods and compositions for preparing noribogaine from voacangine
US8742096B2 (en) 2011-03-28 2014-06-03 Demerx, Inc. Methods and compositions for preparing noribogaine from voacangine
RU2014105881A (en) 2011-09-15 2015-10-20 Демеркс, Инк. NORIBOGAIN SALTS ANSOLVATES
JP6134094B2 (en) 2011-11-04 2017-05-24 株式会社神戸製鋼所 Nitrogen oxide removal method
JP2015509076A (en) 2011-12-08 2015-03-26 デメレックス, インコーポレイテッド Stereoselective total synthesis of nolubogaine.
WO2013085849A2 (en) * 2011-12-09 2013-06-13 Demerx, Inc. Sulfate esters of noribogaine
WO2013085922A1 (en) 2011-12-09 2013-06-13 Demerx, Inc. Phosphate esters of noribogaine
EP2830601A4 (en) * 2012-03-27 2015-09-23 Albany Medical College Blocking of cue-induced drug reinstatement
US9045481B2 (en) * 2012-12-20 2015-06-02 Demerx, Inc. Substituted noribogaine
US8940728B2 (en) * 2012-12-20 2015-01-27 Demerx, Inc. Substituted noribogaine
WO2014098877A1 (en) * 2012-12-20 2014-06-26 Demerx, Inc. Substituted noribogaine
US9561232B2 (en) * 2014-02-18 2017-02-07 Demerx, Inc. Low dose noribogaine for treating nicotine addiction and preventing relapse of nicotine use
US20150258104A1 (en) * 2014-03-13 2015-09-17 Demerx, Inc. Use of noribogaine for the treatment of pain
US20150258106A1 (en) * 2014-03-13 2015-09-17 Demerx, Inc. Methods for acute and long-term treatment of substance abuse
US20150258105A1 (en) * 2014-03-13 2015-09-17 Demerx, Inc. Methods for acute and long-term treatment of alcohol dependence

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3557126A (en) * 1969-08-14 1971-01-19 American Home Prod Derivatives of iboga alkaloids
US6348456B1 (en) * 1994-07-25 2002-02-19 Deborah C. Mash Method of treating chemical dependency in mammals and a composition therefor
US6211360B1 (en) * 1995-08-08 2001-04-03 Albany Medical College Ibogamine congeners
US20030158202A1 (en) * 2001-06-11 2003-08-21 Patrizia Caldirola New compounds
US8362007B1 (en) * 2010-05-11 2013-01-29 Demerx, Inc. Substituted noribogaine

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
LAYER ET AL.: "Structurally modified ibogaine analogs exhibit different affinities for NMDA receptors", EUROPEAN JOURNAL OF PHARMACOLOGY, vol. 309, 1996, pages 159 - 165, XP055261282, Retrieved from the Internet <URL:http://www.if-an.krakow.pl> [retrieved on 20130217] *
See also references of EP2934541A4 *

Cited By (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2018210229A1 (en) * 2017-05-18 2018-11-22 南京明德新药研发股份有限公司 α-7 NICOTINE ACETYLCHOLINE RECEPTOR AGONIST AND APPLICATION THEREOF
US11447510B2 (en) 2017-10-09 2022-09-20 Compass Pathfinder Limited Preparation of psilocybin, different polymorphic forms, intermediates, formulations and their use
US10947257B2 (en) 2017-10-09 2021-03-16 Compass Pathfinder Limited Preparation of psilocybin, different polymorphic forms, intermediates, formulations and their use
US10954259B1 (en) 2017-10-09 2021-03-23 Compass Pathfinder Limited Preparation of psilocybin, different polymorphic forms, intermediates, formulations and their use
US11149044B2 (en) 2017-10-09 2021-10-19 Compass Pathfinder Limited Preparation of psilocybin, different polymorphic forms, intermediates, formulations and their use
US11180517B2 (en) 2017-10-09 2021-11-23 Compass Pathfinder Limited Preparation of psilocybin, different polymorphic forms, intermediates, formulations and their use
US10519175B2 (en) 2017-10-09 2019-12-31 Compass Pathways Limited Preparation of psilocybin, different polymorphic forms, intermediates, formulations and their use
US11505564B2 (en) 2017-10-09 2022-11-22 Compass Pathfinder Limited Preparation of psilocybin, different polymorphic forms, intermediates, formulations and their use
US11629159B2 (en) 2017-10-09 2023-04-18 Compass Pathfinder Limited Preparation of psilocybin, different polymorphic forms, intermediates, formulations and their use
US11851451B2 (en) 2017-10-09 2023-12-26 Compass Pathfinder Limited Preparation of psilocybin, different polymorphic forms, intermediates, formulations and their use
US11939346B2 (en) 2017-10-09 2024-03-26 Compass Pathfinder Limited Preparation of psilocybin, different polymorphic forms, intermediates, formulations and their use
US11564935B2 (en) 2019-04-17 2023-01-31 Compass Pathfinder Limited Method for treating anxiety disorders, headache disorders, and eating disorders with psilocybin
US11738035B2 (en) 2019-04-17 2023-08-29 Compass Pathfinder Limited Method for treating anxiety disorders, headache disorders, and eating disorders with psilocybin

Also Published As

Publication number Publication date
JP2016508979A (en) 2016-03-24
US20150329536A1 (en) 2015-11-19
EP2934541A4 (en) 2016-08-03
US9783535B2 (en) 2017-10-10
EP2934541A1 (en) 2015-10-28
CA2896133A1 (en) 2014-06-26

Similar Documents

Publication Publication Date Title
US9045481B2 (en) Substituted noribogaine
US8940728B2 (en) Substituted noribogaine
US8362007B1 (en) Substituted noribogaine
WO2014098877A1 (en) Substituted noribogaine
JP7248750B2 (en) Opioid receptor ligands and methods of use and preparation thereof
ES2623362T3 (en) Metabolically stable pain relievers and pain medications
US7371875B2 (en) Cytotoxic agents and methods of use
JP4578580B2 (en) Similar compounds of ***e
JP2004520383A (en) Kappa opioid receptor ligand
JP2010511717A (en) Prodrugs and how to create and use them
TW200427690A (en) Opioid and opioid-like compounds and uses thereof
CA3034895A1 (en) Compositions and methods thereof
CN103816152B (en) The Use and preparation method of spectrum of berberine compounds
CA2855990A1 (en) Sulfate esters of noribogaine
Layer et al. Structurally modified ibogaine analogs exhibit differing affinities for NMDA receptors
EP1358177A2 (en) Treatment of affective disorders by the combined action of a nicotinic receptor agonist and a monoaminergic substance
AU2001295436A1 (en) Novel azabicyclooctene derivatives and their medical use
WO2022217859A1 (en) Bisindole alkaloid compound, and synthesis method therefor and use thereof
EP0693070B1 (en) PYRIDO[2,3-b](1,4)BENZODIAZEPINONES AS M2 RECEPTOR LIGAND FOR THE TREATMENT OF NEUROLOGICAL DISORDERS
Akins Design and Synthesis of Salvinorin-Based Compounds for the Opioid Receptors
JPS63303982A (en) Codeine derivative
TW202409032A (en) Fused ring compound, and preparation method therefor and use thereof
Quadri Investigation of the role of alpha7 nicotinic Acetylcholine receptors in inflammatory diseases through the design, synthesis and biological evaluation of new heterocyclic derivatives
WO1993021188A1 (en) Delta opioid receptor-selective benzylidene-substituted morphinans for the treatment of alcohol abuse
WO2021178405A1 (en) Opioid receptor antagonists

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 12890228

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 2015549336

Country of ref document: JP

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 14654503

Country of ref document: US

ENP Entry into the national phase

Ref document number: 2896133

Country of ref document: CA

NENP Non-entry into the national phase

Ref country code: DE

REEP Request for entry into the european phase

Ref document number: 2012890228

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 2012890228

Country of ref document: EP